1
|
Xiong Y, Huang X, Li Y, Nie Y, Yu H, Shi Y, Xue J, Ji Z, Rong K, Zhang X. Integrating larval zebrafish model and network pharmacology for screening and identification of edible herbs with therapeutic potential for MAFLD: A promising drug Smilax glabra Roxb. Food Chem 2025; 464:141470. [PMID: 39406145 DOI: 10.1016/j.foodchem.2024.141470] [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: 08/07/2024] [Revised: 09/17/2024] [Accepted: 09/27/2024] [Indexed: 11/21/2024]
Abstract
Metabolic-associated fatty liver disease (MAFLD) is becoming a prevalent chronic liver disease. Many medicinal and edible herbs exhibit remarkable biological activities in ameliorating MAFLD but lack a comprehensive assessment of their therapeutic efficacy. This study determined total phenolic and flavonoid contents and in vitro antioxidant properties of 34 edible herbs. Smilax glabra Roxb. (SGR), Coreopsis tinctoria Nutt., and Smilax china L. were obtained with the best bioactivity and antioxidant capacity. The high-cholesterol diet-induced larval zebrafish model was established to compare the anti-MAFLD activity of the three herb extracts mentioned above. In vivo experiments revealed that SGR intervention significantly decreased lipid accumulation, alleviated oxidative stress, and modulated intestinal microbiota composition in zebrafish. Furthermore, three potential active components in SGR and their possible mechanisms were explored through network pharmacology and molecular docking. Our study suggested that SGR is a potential candidate for developing new drugs or dietary supplements for MAFLD therapy.
Collapse
Affiliation(s)
- Yinjuan Xiong
- College of Fisheries, Huazhong Agricultural University, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan 430070, China; Hubei Hongshan Laboratory, Wuhan 430070, China
| | - Xixuan Huang
- College of Fisheries, Huazhong Agricultural University, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan 430070, China; Hubei Hongshan Laboratory, Wuhan 430070, China
| | - Yuxin Li
- College of Fisheries, Huazhong Agricultural University, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan 430070, China; Hubei Hongshan Laboratory, Wuhan 430070, China
| | - Yukang Nie
- College of Fisheries, Huazhong Agricultural University, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan 430070, China; Hubei Hongshan Laboratory, Wuhan 430070, China
| | - Haodong Yu
- College of Fisheries, Huazhong Agricultural University, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan 430070, China; Hubei Hongshan Laboratory, Wuhan 430070, China
| | - Yaqi Shi
- College of Fisheries, Huazhong Agricultural University, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan 430070, China; Hubei Hongshan Laboratory, Wuhan 430070, China
| | - Jiajie Xue
- College of Fisheries, Huazhong Agricultural University, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan 430070, China; Hubei Hongshan Laboratory, Wuhan 430070, China
| | - Zhehui Ji
- College of Fisheries, Huazhong Agricultural University, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan 430070, China; Hubei Hongshan Laboratory, Wuhan 430070, China
| | - Keming Rong
- Research Institute of Huanong-Tianchen, Wuhan 430070, China
| | - Xuezhen Zhang
- College of Fisheries, Huazhong Agricultural University, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Hubei Provincial Engineering Laboratory for Pond Aquaculture, Wuhan 430070, China; Hubei Hongshan Laboratory, Wuhan 430070, China; Research Institute of Huanong-Tianchen, Wuhan 430070, China.
| |
Collapse
|
2
|
Luo X, Fang Y, Wang W, Tong M, Qin B, Cao J, Yang Y. Yinchen lipid-lowering tea attenuates lipid deposition in a fatty liver model by regulating mitochondrial dysfunction through activation of AdipoR1/AMPK/SIRT1 signaling. 3 Biotech 2025; 15:39. [PMID: 39807243 PMCID: PMC11725549 DOI: 10.1007/s13205-024-04204-2] [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: 07/11/2024] [Accepted: 12/29/2024] [Indexed: 01/16/2025] Open
Abstract
This study investigated the ameliorative effects of Yinchen lipid-lowering tea (YCLLT) on Non-alcoholic fatty liver disease (NAFLD), the specific mechanism involved was also studied. We modeled hepatocellular steatosis with HepG2 cells and intervened with different concentrations of YCLLT-containing serum. Lipid deposition was assessed by oil red O staining and AdipoR1 expression was analyzed by Western blot. The hepatocyte steatosis model was further treated with YCLLT-containing serum and/or silencing AdipoR1. Lipid deposition was observed by oil red O staining. Flow cytometry was used to detect apoptosis and mitochondrial membrane potential. The levels of TNF-α, IL-6, MDA, 8-OHdG, and ATP were analyzed by ELISA or the corresponding kits. The mitochondrial structure was observed by transmission electron microscopy. The expression of AdipoR1/AMPK/SIRT1 signaling pathway factors was analyzed by Western blot, and co-localization of SIRT1 and immunofluorescence. The results revealed that YCLLT attenuated lipid deposition, inhibited the levels of inflammatory factors TNF-α and IL-6, reduced the levels of MDA and 8-OHdG, up-regulated the ATP content and mitochondrial membrane potential, and promoted the expression of AdipoR1, p-LKB1, p-AMPKα, SIRT1, and PGC-1a in a cellular model of NAFLD. Further, silencing of AdipoR1 inhibited the ameliorative effect of YCLLT in the NAFLD cell model. Altogether, Yinchen lipid-lowering tea attenuates lipid deposition in a fatty liver model by improving mitochondrial function via activating AdipoR1/AMPK/ SIRT1 signaling.
Collapse
Affiliation(s)
- Xilin Luo
- Department of Preventive Treatment of Disease Centre, Nanchong Chinese Medicine Hospital (Nanchong Traditional Chinese Medicine Hospital Affiliated to North Sichuan Medical College), 200 Jingyuling Zhengjie Road, Shunqing District, Nanchong City, Sichuan Province 637000 People’s Republic of China
| | - Yuanyuan Fang
- Department of Preventive Treatment of Disease Centre, Nanchong Chinese Medicine Hospital (Nanchong Traditional Chinese Medicine Hospital Affiliated to North Sichuan Medical College), 200 Jingyuling Zhengjie Road, Shunqing District, Nanchong City, Sichuan Province 637000 People’s Republic of China
| | - Wei Wang
- Department of Preventive Treatment of Disease Centre, Nanchong Chinese Medicine Hospital (Nanchong Traditional Chinese Medicine Hospital Affiliated to North Sichuan Medical College), 200 Jingyuling Zhengjie Road, Shunqing District, Nanchong City, Sichuan Province 637000 People’s Republic of China
| | - Meiling Tong
- Department of Preventive Treatment of Disease Centre, Nanchong Chinese Medicine Hospital (Nanchong Traditional Chinese Medicine Hospital Affiliated to North Sichuan Medical College), 200 Jingyuling Zhengjie Road, Shunqing District, Nanchong City, Sichuan Province 637000 People’s Republic of China
| | - Bin Qin
- Department of Preventive Treatment of Disease Centre, Nanchong Chinese Medicine Hospital (Nanchong Traditional Chinese Medicine Hospital Affiliated to North Sichuan Medical College), 200 Jingyuling Zhengjie Road, Shunqing District, Nanchong City, Sichuan Province 637000 People’s Republic of China
| | - Jinyu Cao
- Department of Preventive Treatment of Disease Centre, Nanchong Chinese Medicine Hospital (Nanchong Traditional Chinese Medicine Hospital Affiliated to North Sichuan Medical College), 200 Jingyuling Zhengjie Road, Shunqing District, Nanchong City, Sichuan Province 637000 People’s Republic of China
| | - Yinjie Yang
- Department of Preventive Treatment of Disease Centre, Nanchong Chinese Medicine Hospital (Nanchong Traditional Chinese Medicine Hospital Affiliated to North Sichuan Medical College), 200 Jingyuling Zhengjie Road, Shunqing District, Nanchong City, Sichuan Province 637000 People’s Republic of China
| |
Collapse
|
3
|
He Y, Ye M, Xia Y, Zhong Z, Li Q. Antioxidants and the risk of metabolic dysfunction-associated steatotic liver disease: results of National Health and Nutrition Examination Survey and two-sample Mendelian randomization analyses. Eur J Gastroenterol Hepatol 2025; 37:230-239. [PMID: 39621882 DOI: 10.1097/meg.0000000000002898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2024]
Abstract
BACKGROUND The link between antioxidants and metabolic dysfunction-associated steatotic liver disease (MASLD) is a topic of considerable discussion in the field of observational studies, with the exact causal connections still being unclear. METHODS In this investigation, a cohort consisting of 17 061 participants from the National Health and Nutrition Examination Surveys was studied. Initially, a cross-sectional analysis was carried out to examine the relationship between the CDAI and MASLD. Further, Mendelian randomization (MR) was utilized to assess the possible causal links between antioxidant levels in the bloodstream and MASLD. RESULTS The association between the CDAI and MASLD was found to be significant in the fully adjusted logistic regression model, showing an OR of 0.95 [95% confidence interval (CI): 0.94-0.97; P < 0.001]. The use of restricted cubic spline regression revealed no significant nonlinear association between the CDAI and the occurrence of MASLD ( Pnonlinearity = 0.321). Additionally, MR findings did not suggest any causal connections between circulating levels of various antioxidants and MASLD. These antioxidants included vitamin A (retinol) (IVW: OR: 0.67, 95% CI: 0.33-1.36, P = 0.272), vitamin C (ascorbate) (IVW: OR: 0.61, 95% CI: 0.34-1.09, P = 0.094), vitamin E (α-tocopherol) (IVW: OR: 0.55, 95% CI: 0.13-2.25, P = 0.407), vitamin E (γ-tocopherol) (IVW: OR: 0.89, 95% CI: 0.36-2.23, P = 0.806), zinc (IVW: OR: 0.95, 95% CI: 0.82-1.09, P = 0.449), selenium (IVW: OR: 0.98, 95% CI: 0.84-1.16, P = 0.855), and carotene (IVW: OR: 0.80, 95% CI: 0.36-1.81, P = 0.596). CONCLUSION The findings highlight a significant negative linear relationship between CDAI and MASLD prevalence in the observational component of the study. However, the MR analysis did not indicate any causal effects of circulating antioxidant levels on MASLD.
Collapse
Affiliation(s)
- Yijia He
- Department of Endocrinology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | | | | | | | | |
Collapse
|
4
|
Ding T, Shen W, Tao W, Peng J, Pan M, Qi X, Feng W, Wei N, Zheng S, Jin H. Curcumol ameliorates alcohol and high-fat diet-induced fatty liver disease via modulation of the Ceruloplasmin/iron overload/mtDNA signaling pathway. J Nutr Biochem 2025; 136:109807. [PMID: 39549858 DOI: 10.1016/j.jnutbio.2024.109807] [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: 07/17/2024] [Revised: 09/28/2024] [Accepted: 11/11/2024] [Indexed: 11/18/2024]
Abstract
Fatty liver disease (FLD), a chronic liver disease characterized by excessive lipid deposition, is affecting more and more people worldwide owing to the increasing global incidence of obesity and heavy alcohol consumption. However, there is still no effective strategy for prevention or treatment of alcohol and high-fat diet (HFD)-induced FLD. The purpose of this study was to investigate the effect of curcumol on alcohol and HFD-induced FLD and the underlying molecular mechanisms. The results showed that curcumol ameliorated alcohol and HFD-induced hepatocyte injury in vivo and in vitro, and the mechanism might be related to its up-regulation of ceruloplasmin and subsequent alleviation of iron overload. Moreover, curcumol inhibited alcohol and HFD-induced mitochondrial damage and mtDNA release in hepatocytes by modulating iron overload. Furthermore, curcumol's inhibition of mtDNA release could suppress the activation of cGAS-STING and subsequent inflammation, and this phenomenon could be reversed by cGAS overexpression. Notably, alcohol and HFD-induced mtDNA release from hepatocytes contributed to HSC activation and this effect could be weakened by curcumol. In conclusion, these findings elucidated that curcumol ameliorated alcohol and HFD-induced FLD via modulating ceruloplasmin/iron overload/mtDNA signaling pathway, which lead to the inhibition of inflammation and HSCs activation.
Collapse
Affiliation(s)
- Tingting Ding
- Department of Pharmacology, School of Pharmacy, Wannan Medical College, Wuhu, Anhui, China
| | - Wanqing Shen
- Department of Pharmacology, School of Pharmacy, Wannan Medical College, Wuhu, Anhui, China
| | - Wenhui Tao
- Department of Pharmacology, School of Pharmacy, Wannan Medical College, Wuhu, Anhui, China
| | - Junlu Peng
- Department of Digestive surgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui, China
| | - Meijun Pan
- Department of Pharmacology, School of Pharmacy, Wannan Medical College, Wuhu, Anhui, China
| | - Xiaoyu Qi
- Department of Pharmacy, The Second Affiliated Hospital of Wannan Medical College, Wuhu, Anhui, China
| | - Wanyu Feng
- Department of Pharmacology, School of Pharmacy, Wannan Medical College, Wuhu, Anhui, China
| | - Na Wei
- Department of Pharmacology, School of Pharmacy, Wannan Medical College, Wuhu, Anhui, China
| | - Shuguo Zheng
- Department of Pharmacology, School of Pharmacy, Wannan Medical College, Wuhu, Anhui, China; Laboratory of Pharmacology of Chinese Medicine, School of Pharmacy, Wannan Medical College, Wuhu, Anhui, China.
| | - Huanhuan Jin
- Department of Pharmacology, School of Pharmacy, Wannan Medical College, Wuhu, Anhui, China; Laboratory of Pharmacology of Chinese Medicine, School of Pharmacy, Wannan Medical College, Wuhu, Anhui, China.
| |
Collapse
|
5
|
Gerardo H, Oliveira PJ, Cavadas C, Grãos M, Teixeira J. The (un)known crosstalk between metabolism and mechanotransduction: Implications for metabolic syndrome (MetS)-associated neurological complications. Biochim Biophys Acta Mol Basis Dis 2025:167678. [PMID: 39832691 DOI: 10.1016/j.bbadis.2025.167678] [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: 09/30/2024] [Revised: 01/15/2025] [Accepted: 01/15/2025] [Indexed: 01/22/2025]
Abstract
Metabolic syndrome (MetS) has been associated with disruptions in tissue mechanical homeostasis and inflammatory and metabolic derangements. However, the direct correlation between metabolic alterations and changes in tissue stiffness, and whether they could play a role as upstream initiators of disease pathology remains to be investigated. This emerging concept has yet to be put into clinical practice as many questions concerning the interplay between extracellular matrix mechanical properties and regulation of metabolic pathways remain unsolved. This review will highlight key foundational studies examining mutual regulation of cell metabolism and mechanotransduction, and opening questions lying ahead for better understanding MetS pathophysiology.
Collapse
Affiliation(s)
- Heloísa Gerardo
- CNC-UC, Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; CIBB, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal.
| | - Paulo J Oliveira
- CNC-UC, Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; CIBB, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Portugal.
| | - Cláudia Cavadas
- CNC-UC, Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; CIBB, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal.
| | - Mário Grãos
- CIBB, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Portugal
| | - José Teixeira
- CNC-UC, Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; CIBB, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Portugal.
| |
Collapse
|
6
|
Zhou M, Lv J, Chen X, Shi Y, Chao G, Zhang S. From gut to liver: Exploring the crosstalk between gut-liver axis and oxidative stress in metabolic dysfunction-associated steatotic liver disease. Ann Hepatol 2025:101777. [PMID: 39832564 DOI: 10.1016/j.aohep.2025.101777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 01/05/2025] [Accepted: 01/10/2025] [Indexed: 01/22/2025]
Abstract
Non-alcoholic fatty liver disease (NAFLD), now recognized as metabolic dysfunction-associated steatotic liver disease (MASLD), represents a significant and escalating global health challenge. Its prevalence is intricately linked to obesity, insulin resistance, and other components of the metabolic syndrome. As our comprehension of MASLD deepens, it has become evident that this condition extends beyond the liver, embodying a complex, multi-systemic disease with hepatic manifestations that mirror the broader metabolic landscape. This comprehensive review delves into the critical interplay between the gut-liver axis and oxidative stress, elucidating their pivotal roles in the etiology and progression of MASLD. Our analysis reveals several key findings: (1) Bile acid dysregulation can trigger oxidative stress through enhanced ROS production in hepatocytes and Kupffer cells, leading to mitochondrial dysfunction and lipid peroxidation; (2) Gut microbiota dysbiosis disrupts intestinal barrier function, allowing increased translocation of endotoxins like LPS, which activate inflammatory pathways through TLR4 signaling and promote oxidative stress via NADPH oxidase activation; (3) The redox-sensitive transcription factors NF-κB and Nrf2 serve as crucial mediators in the gut-liver axis, with NF-κB regulating inflammatory responses and Nrf2 orchestrating antioxidant defenses; (4) Oxidative stress-induced damage to intestinal barrier function creates a destructive feedback loop, further exacerbating liver inflammation and disease progression. These findings highlight the complex interrelationship between gut-liver axis dysfunction and oxidative stress in MASLD pathogenesis, suggesting potential therapeutic targets for disease management.
Collapse
Affiliation(s)
- Mi Zhou
- Department of Gastroenterology, Xinhua Hospital of zhejiang Province: The Second Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang, China
| | - Jianyu Lv
- Department of Gastroenterology, Xinhua Hospital of zhejiang Province: The Second Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang, China
| | - Xinli Chen
- Department of Gastroenterology, Xinhua Hospital of zhejiang Province: The Second Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang, China
| | - Yujie Shi
- Department of Gastroenterology, Xinhua Hospital of zhejiang Province: The Second Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang, China
| | - Guanqun Chao
- Department of General Practice, Zhejiang University School of Medicine Sir Run Run Shaw Hospital, China
| | - Shuo Zhang
- Department of Gastroenterology, Xinhua Hospital of zhejiang Province: The Second Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang, China.
| |
Collapse
|
7
|
Zhang S, Wang N, Gao Z, Gao J, Wang X, Xie H, Wang CY, Zhang S. Reductive stress: The key pathway in metabolic disorders induced by overnutrition. J Adv Res 2025:S2090-1232(25)00031-1. [PMID: 39805424 DOI: 10.1016/j.jare.2025.01.012] [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: 09/01/2024] [Revised: 12/04/2024] [Accepted: 01/05/2025] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND The balance of redox states is crucial for maintaining physiological homeostasis. For decades, the focus has been mainly on the concept of oxidative stress, which is involved in the mechanism of almost all diseases. However, robust evidence has highlighted that reductive stress, the other side of the redox spectrum, plays a pivotal role in the development of various diseases, particularly those related to metabolism and cardiovascular health. AIM OF REVIEW In this review, we present an extensive array of evidence for the occurrence of reductive stress and its significant implications mainly in metabolic and cardiovascular diseases. KEY SCIENTIFIC CONCEPTS OF REVIEW Reductive stress is defined as a shift in the cellular redox balance towards a more reduced state, characterized by an excess of endogenous reductants (such as NADH, NADPH, and GSH) over their oxidized counterparts (NAD+, NADP+, and GSSG). While oxidative stress has been the predominant mechanism studied in obesity, metabolic disorders, and cardiovascular diseases, growing evidence underscores the critical role of reductive stress. This review discusses how reductive stress contributes to metabolic and cardiovascular pathologies, emphasizing its effects on key cellular processes. For example, excessive NADH accumulation can disrupt mitochondrial function by impairing the electron transport chain, leading to decreased ATP production and increased production of reactive oxygen species. In the endoplasmic reticulum (ER), an excess of reductive equivalents hampers protein folding, triggering ER stress and activating the unfolded protein response, which can lead to insulin resistance and compromised cellular homeostasis. Furthermore, we explore how excessive antioxidant supplementation can exacerbate reductive stress by further shifting the redox balance, potentially undermining the beneficial effects of exercise, impairing cardiovascular health, and aggravating metabolic disorders, particularly in obese individuals. This growing body of evidence calls for a reevaluation of the role of reductive stress in disease pathogenesis and therapeutic interventions.
Collapse
Affiliation(s)
- Shiyi Zhang
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Na Wang
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhichao Gao
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia Gao
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaohui Wang
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hao Xie
- Institute of Translational Medicine, Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.
| | - Cong-Yi Wang
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Shu Zhang
- The Center for Biomedical Research, Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
8
|
Yin W, Xu H, Bai Z, Wu Y, Zhang Y, Liu R, Wang Z, Zhang B, Shen J, Zhang H, Chen X, Ma D, Shi X, Yan L, Zhang C, Jiang H, Chen K, Guo D, Niu W, Yin H, Zhang WJ, Luo C, Xie X. Inhibited peroxidase activity of peroxiredoxin 1 by palmitic acid exacerbates nonalcoholic steatohepatitis in male mice. Nat Commun 2025; 16:598. [PMID: 39799115 PMCID: PMC11724923 DOI: 10.1038/s41467-025-55939-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 01/06/2025] [Indexed: 01/15/2025] Open
Abstract
Reactive oxygen species exacerbate nonalcoholic steatohepatitis (NASH) by oxidizing macromolecules; yet how they promote NASH remains poorly understood. Here, we show that peroxidase activity of global hepatic peroxiredoxin (PRDX) is significantly decreased in NASH, and palmitic acid (PA) binds to PRDX1 and inhibits its peroxidase activity. Using three genetic models, we demonstrate that hepatic PRDX1 protects against NASH in male mice. Mechanistically, PRDX1 suppresses STAT signaling and protects mitochondrial function by scavenging hydrogen peroxide, and mitigating the oxidation of protein tyrosine phosphatases and lipid peroxidation. We further identify rosmarinic acid (RA) as a potent agonist of PRDX1. As revealed by the complex crystal structure, RA binds to PRDX1 and stabilizes its peroxidatic cysteine. RA alleviates NASH through specifically activating PRDX1's peroxidase activity. Thus, beyond revealing the molecular mechanism underlying PA promoting oxidative stress and NASH, our study suggests that boosting PRDX1's peroxidase activity is a promising intervention for treating NASH.
Collapse
Affiliation(s)
- Wen Yin
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Heng Xu
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310000, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Zhonghao Bai
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
- Department of Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), NHC Key Laboratory of Hormones and Development, Tianjin Medical University, Tianjin, 300070, China
| | - Yue Wu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing, 100049, China
| | - Yan Zhang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Rui Liu
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Zhangzhao Wang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Bei Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing, 100049, China
| | - Jing Shen
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Hao Zhang
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310000, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xin Chen
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety Research, Shanghai Institute of Nutrition and Health (SINH), Chinese Academy of Sciences (CAS), Shanghai, China
| | - Danting Ma
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Xiaofeng Shi
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Lihui Yan
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Chang Zhang
- Department of Pharmacy, General Hospital, Tianjin Medical University, Tianjin, 300070, China
| | - Hualiang Jiang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Kaixian Chen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Dean Guo
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Wenyan Niu
- Department of Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), NHC Key Laboratory of Hormones and Development, Tianjin Medical University, Tianjin, 300070, China
| | - Huiyong Yin
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety Research, Shanghai Institute of Nutrition and Health (SINH), Chinese Academy of Sciences (CAS), Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China
| | - Weiping J Zhang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Cheng Luo
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310000, China.
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing, 100049, China.
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Xiangyang Xie
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, The province and ministry co-sponsored collaborative innovation center for medical epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China.
| |
Collapse
|
9
|
Liu B, Shao T, Xiao D, Yang S, Lin W, Sun L, Zhang W, Luo M, Zhao J, Yang L, Bai S, Deng D, Wang C, Wang S, Zhang R, Liu Z, An L. Aqueous extract of Cornus officinalis alleviate NAFLD via protecting hepatocytes proliferation through regulation of the tricarboxylic acid cycle. JOURNAL OF ETHNOPHARMACOLOGY 2025; 341:119330. [PMID: 39778783 DOI: 10.1016/j.jep.2025.119330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 12/02/2024] [Accepted: 01/05/2025] [Indexed: 01/11/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cornus officinalis (CO) has been widely used as Chinese herbal medicine and has a good clinical efficacy in liver disease. In particular, it has a significant therapeutic effect on metabolic liver disease. However, systematic pharmacological studies on its hepatoprotective effect on non-alcoholic fatty liver disease (NAFLD) are lacking. AIM OF THE STUDY We investigated the impact of Cornus officinalis extract (COE) on two mouse models of NAFLD, screened the potential mechanisms of action by using metabolomics assays, and explored the protective effects on hepatocyte proliferation by regulating glutamate metabolism and tricarboxylic acid (TCA) cycle. METHODS The main components of COE were identified by high performance liquid chromatograph (HPLC). Male C57BL/6J mice were subjected to construct carbon tetrachloride (CCl4) or methionine choline deficient (MCD) induced NAFLD mice. Liver function and lipid biochemical indicators were detected using commercial assay kits. Masson staining, Western blot, and immunohistochemistry analyses were used for assessing hepatic injury and fibrosis. LC-MS non-targeted analysis was performed using the 1290 Ultra-High Performance Liquid Chromatograph System and the 6540 Q-TOF Mass Spectrometry. Palmitic acid (PA) induced L-02 cell model was established. The mediators in glutamate metabolism and TCA cycle were assessed by assay kits. RESULTS In vivo experiments validated that COE significantly improved liver function in NAFLD mice, reduced lipid accumulation, and alleviated pathological damage and liver fibrosis. The non-targeted metabolomics analysis combined with Ingenuity Pathway Analysis (IPA) located glutamate metabolism and the downstream TCA cycle as potential mechanisms of COE, which was further confirmed in NAFLD model mice and PA-induced L-02 cells. Finally, we confirmed that COE could promote mitochondrial energy supply by remodeling the homeostasis of the TCA cycle, thereby enhancing hepatocyte proliferation. CONCLUSIONS This study demonstrated that COE could significantly improve CCl4 or MCD-induced NAFLD by promoting hepatocyte proliferation. These results highlighted the potential of COE as leads for the development of innovative treatments for NAFLD.
Collapse
Affiliation(s)
- Binjie Liu
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Ting Shao
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Dandan Xiao
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Chinese Medicine Guangdong Laboratory, Hengqin, Guangdong, China
| | - Shujie Yang
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Chinese Medicine Guangdong Laboratory, Hengqin, Guangdong, China
| | - Weijie Lin
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Lizhu Sun
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Weiqin Zhang
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Meiqing Luo
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jinlan Zhao
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Lei Yang
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Shasha Bai
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Chinese Medicine Guangdong Laboratory, Hengqin, Guangdong, China
| | - Di Deng
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Chinese Medicine Guangdong Laboratory, Hengqin, Guangdong, China
| | - Caiyan Wang
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Chinese Medicine Guangdong Laboratory, Hengqin, Guangdong, China; State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou, Guangdong, China
| | - Shaogui Wang
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Rong Zhang
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Chinese Medicine Guangdong Laboratory, Hengqin, Guangdong, China; State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou, Guangdong, China.
| | - Zhongqiu Liu
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Chinese Medicine Guangdong Laboratory, Hengqin, Guangdong, China; State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou, Guangdong, China.
| | - Lin An
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; Chinese Medicine Guangdong Laboratory, Hengqin, Guangdong, China.
| |
Collapse
|
10
|
Srinivasan D, Subbarayan R, Krishnan M, Balakrishna R, Adtani P, Shrestha R, Chauhan A, Babu S, Radhakrishnan A. Radiation therapy-induced normal tissue damage: involvement of EMT pathways and role of FLASH-RT in reducing toxicities. RADIATION AND ENVIRONMENTAL BIOPHYSICS 2025:10.1007/s00411-024-01102-2. [PMID: 39760753 DOI: 10.1007/s00411-024-01102-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 12/20/2024] [Indexed: 01/07/2025]
Abstract
Radiation therapy (RT) is fundamental to the fight against cancer because of its exceptional ability to target and destroy cancer cells. However, conventional radiation therapy can significantly affect the adjacent normal tissues, leading to fibrosis, inflammation, and decreased organ function. This tissue damage not only reduces the quality of life but also prevents the total elimination of cancer. The transformation of epithelial cells into mesenchymal-like cells, termed epithelial-mesenchymal transition (EMT), is essential for processes such as fibrosis, embryogenesis, and wound healing. Conventional radiation therapy increases the asymmetric activation of fibrotic and inflammatory pathways, and the resulting chronic fibrotic changes and organ dysfunction are linked to radiation-induced epithelial-mesenchymal transition. Recent advances in radiation therapy, namely flash radiation therapy (FLASH-RT), have the potential to widen the therapeutic index. Radiation delivered by FLASH-RT at very high dose rates (exceeding 40 Gy/s) can protect normal tissue from radiation-induced damage, a phenomenon referred to as the "FLASH effect". Preclinical studies have demonstrated that FLASH-RT successfully inhibits processes associated with fibrosis and epithelial-mesenchymal transition, mitigates damage to normal tissue, and enhances regeneration. Three distinct types of EMT have been identified: type-1, associated with embryogenesis; Type-2, associated with injury potential; and type-3, related with cancer spread. The regulation of EMT via pathways, including TGF-β/SMAD, WNT/β-catenin, and NF-κB, is essential for radiation-induced tissue remodelling. This study examined radiation-induced EMT, TGF-β activity, multiple signalling pathways in fibrosis, and the potential of FLASH-RT to reduce tissue damage. FLASH-RT is a novel approach to treat chronic tissue injury and fibrosis post-irradiation by maintaining epithelial properties and regulating mesenchymal markers including vimentin and N-cadherin. Understanding these pathways will facilitate the development of future therapies that can alleviate fibrosis, improve the efficacy of cancer therapy, and improve the quality of life of patients.
Collapse
Affiliation(s)
- Dhasarathdev Srinivasan
- Centre for Advanced Biotherapeutics and Regenerative Medicine, Faculty of Research, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, 603103, Tamil Nadu, India
| | - Rajasekaran Subbarayan
- Centre for Advanced Biotherapeutics and Regenerative Medicine, Faculty of Research, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, 603103, Tamil Nadu, India.
- Centre for Herbal Pharmacology and Environmental Sustainability, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, 603103, Tamil Nadu, India.
| | - Madhan Krishnan
- Faculty of Research, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, 603103, Tamil Nadu, India
| | - Ranjith Balakrishna
- Centre for Advanced Biotherapeutics and Regenerative Medicine, Faculty of Research, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, 603103, Tamil Nadu, India
| | - Pooja Adtani
- Department of Basic Medical and Dental Sciences, Gulf Medical University, Ajman, United Arab Emirates
| | - Rupendra Shrestha
- Department of Natural and Applied Sciences, Nexus Institute of Research and Innovation (NIRI), Lalitpur, Nepal.
| | - Ankush Chauhan
- Centre for Herbal Pharmacology and Environmental Sustainability, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, 603103, Tamil Nadu, India
| | - Shyamaladevi Babu
- Faculty of Research, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, 603103, Tamil Nadu, India
| | - Arunkumar Radhakrishnan
- Department of Pharmacology, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, India
| |
Collapse
|
11
|
de Calbiac H, Imbard A, de Lonlay P. Cellular mechanisms of acute rhabdomyolysis in inherited metabolic diseases. J Inherit Metab Dis 2025; 48:e12781. [PMID: 39135340 DOI: 10.1002/jimd.12781] [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: 01/09/2024] [Revised: 07/02/2024] [Accepted: 07/04/2024] [Indexed: 12/28/2024]
Abstract
Acute rhabdomyolysis (RM) constitutes a life-threatening emergency resulting from the (acute) breakdown of skeletal myofibers, characterized by a plasma creatine kinase (CK) level exceeding 1000 IU/L in response to a precipitating factor. Genetic predisposition, particularly inherited metabolic diseases, often underlie RM, contributing to recurrent episodes. Both sporadic and congenital forms of RM share common triggers. Considering the skeletal muscle's urgent need to rapidly adjust to environmental cues, sustaining sufficient energy levels and functional autophagy and mitophagy processes are vital for its preservation and response to stressors. Crucially, the composition of membrane lipids, along with lipid and calcium transport, and the availability of adenosine triphosphate (ATP), influence membrane biophysical properties, membrane curvature in skeletal muscle, calcium channel signaling regulation, and determine the characteristics of autophagic organelles. Consequently, a genetic defect involving ATP depletion, aberrant calcium release, abnormal lipid metabolism and/or lipid or calcium transport, and/or impaired anterograde trafficking may disrupt autophagy resulting in RM. The complex composition of lipid membranes also alters Toll-like receptor signaling and viral replication. In response, infections, recognized triggers of RM, stimulate increased levels of inflammatory cytokines, affecting skeletal muscle integrity, energy metabolism, and cellular trafficking, while elevated temperatures can reduce the activity of thermolabile enzymes. Overall, several mechanisms can account for RMs and may be associated in the same disease-causing RM.
Collapse
Affiliation(s)
- Hortense de Calbiac
- INSERM U1151, Institut Necker Enfants-Malades (INEM), Université Paris Cité, Paris, France
| | - Apolline Imbard
- Service de Biochimie, Hôpital Universitaire Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
- Faculté de pharmacie, LYPSIS, Université Paris Saclay, Orsay, France
- Reference Center for Inherited Metabolic Diseases, Necker-Enfants-Malades University Hospital, APHP, Imagine Institute, Filière G2M, MetabERN, Paris, France
| | - Pascale de Lonlay
- INSERM U1151, Institut Necker Enfants-Malades (INEM), Université Paris Cité, Paris, France
- Reference Center for Inherited Metabolic Diseases, Necker-Enfants-Malades University Hospital, APHP, Imagine Institute, Filière G2M, MetabERN, Paris, France
| |
Collapse
|
12
|
Zeng W, Wang L, Wang C, Xiong X, Huang Q, Chen S, Liu C, Liu W, Wang Y, Huang Q. SENP1 prevents high fat diet-induced non-alcoholic fatty liver diseases by regulating mitochondrial dynamics. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167527. [PMID: 39332783 DOI: 10.1016/j.bbadis.2024.167527] [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: 04/06/2024] [Revised: 09/12/2024] [Accepted: 09/22/2024] [Indexed: 09/29/2024]
Abstract
Mitochondrial dynamics plays a crucial role in the occurrence and development of non-alcoholic fatty liver diseases (NAFLD). SENP1, a SUMO-specific protease, catalyzes protein de-SUMOylation and involves in various physiological and pathological processes. However, the exact role of SENP1 in NAFLD remains unclear. Therefore, we investigated the regulatory role of SENP1 in mitochondrial dynamics during the progression of NAFLD. In the study, the NAFLD in vivo model induced by high fat diet (HFD) and in vitro model induced by free fatty acids (FFA) were established to investigate the role and underlying mechanism of SENP1 through detecting mitochondrial morphology and dynamics. Our results showed that the down-regulation of SENP1 expression and the mitochondrial dynamics dysregulation occurred in the NAFLD, evidenced as mitochondrial fragmentation, up-regulation of p-Drp1 ser616 and down-regulation of MFN2, OPA1. However, over-expression of SENP1 significantly alleviated the NAFLD, rectified the mitochondrial dynamics disorder, reduced Cyt-c release and ROS levels induced by FFA or HFD; moreover, the over-expression of SENP1 also reduced the SUMOylation levels of Drp1 and prevented the Drp1 translocation to mitochondria. Our findings suggest that the possible mechanisms of SENP1 were through rectifying the mitochondrial dynamics disorder, reducing Cyt-c release and ROS-mediated oxidative stress. The findings would provide a novel target for the prevention and treatment of NALFD.
Collapse
Affiliation(s)
- Wenjing Zeng
- Department of Pharmacology, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, PR China; Provincial Key Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Li Wang
- Department of Pharmacology, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, PR China; Provincial Key Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Chaowen Wang
- Department of Pharmacology, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, PR China; Provincial Key Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Xiaowei Xiong
- Department of Pharmacology, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, PR China; Provincial Key Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Qianqian Huang
- Department of Pharmacology, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, PR China; Provincial Key Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Sheng Chen
- Department of Pharmacology, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, PR China; Provincial Key Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Chen Liu
- Department of Pharmacology, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, PR China; Provincial Key Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Wentao Liu
- Department of Pharmacology, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, PR China; Provincial Key Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Yuan Wang
- Department of Pharmacology, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, PR China; Provincial Key Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Qiren Huang
- Department of Pharmacology, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, PR China; Provincial Key Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, 330006, PR China.
| |
Collapse
|
13
|
Li Y, Duan X, Wang Y, Deng Y, Zhang J. Structural characterization and in vitro hepatoprotective activity of an acidic polysaccharide from Dendrobium nobile Lindl. flower. Int J Biol Macromol 2025; 284:138100. [PMID: 39608518 DOI: 10.1016/j.ijbiomac.2024.138100] [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: 06/06/2024] [Revised: 11/02/2024] [Accepted: 11/25/2024] [Indexed: 11/30/2024]
Abstract
This study aimed to investigate the structural characterization, conformational properties, and hepatoprotectiveactivity of acidic polysaccharide (DNLPS-3) from Dendrobium nobile flower. DNLPS-3 with molecular weights of 15.65 KDa was composed of Fuc: Rha: Ara: Gal: Glc: Xyl: GalA: GlcA in a molar ratio of 0.26%: 2.38%: 4.47%: 3.55%: 0.43%: 17.84%: 69.80%: 1.26%. Structural analysis with methylation indicated that DNLPS-3 mainly contained T-Ara(f), T-Xyl(p), T-Gal(p)A, 3-Gal(p)A, 4-Gal(p)A, 3,4-Gal(p)A, 3,4-Glc(p), 2,4-Gal(p)A, 2,3,4-Gal(p), 4,6-Gal(p), 3,4,6-Gal(p). NMR results show that DNLPS-3 may be a pectin polysaccharide with →4)-α-Gal(p)A-(→main chain. In vitro experiments showed that DNLPS-3 exhibited significant hepatoprotective effects. The effect was achieved by reducing the activities of ALT and AST, suppressing the production of LDH and MDA, and enhancing the anti-oxidant activities of SOD and GSH, and the specific mechanism of its hepatoprotective effect is that DNLPS-3 up-regulates Nrf2/HO-1 pathway protein expression.
Collapse
Affiliation(s)
- Yao Li
- School of pharmacy, Zunyi Medical University, Zunyi 563000, China
| | - Xiaotong Duan
- School of pharmacy, Zunyi Medical University, Zunyi 563000, China
| | - Yueyue Wang
- Hospital of Stomatology, Zunyi Medical University, Zunyi 563000, China
| | - Yong Deng
- School of pharmacy, Zunyi Medical University, Zunyi 563000, China; State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Beijing 100700, China.
| | - Jianyong Zhang
- School of Pharmacy and Key Laboratory of Basic Pharmacology Ministry Education and Joint International Research Laboratory of Ethnomedicine Ministry of Education, Zunyi Medical University, Zunyi 563000, China.
| |
Collapse
|
14
|
Ha YS, Kim TK, Heo J, Oh J, Kim SK, Kim J, Lee J, Yang SR, Hwang S, Kim SJ. Rocaglamide-A mitigates LPS-induced hepatic inflammation by modulating JNK/AP-1 signaling cascade and ROS production in hepatocytes. Toxicol Res 2025; 41:47-59. [PMID: 39802115 PMCID: PMC11717754 DOI: 10.1007/s43188-024-00263-y] [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: 06/04/2024] [Revised: 08/23/2024] [Accepted: 09/10/2024] [Indexed: 01/03/2025] Open
Abstract
Lipopolysaccharide (LPS), a gut-derived endotoxin, is a recognized risk factor for both Non-alcoholic fatty liver disease (NAFLD) and alcoholic liver disease (ALD). Rocaglamide-A (Roc-A), a natural compound derived from the genus Aglaia, is known for its pharmacological and immunosuppressive effects on various cell types. Although our recent investigations have unveiled Roc-A's anti-adipogenic role in adipocytes, its mechanism in hepatic inflammation remains elusive. This study delves into Roc-A's protective effects on LPS-induced hepatic inflammation. Our results demonstrated that Roc-A treatment significantly reduced the LPS-induced production of inflammatory cytokines in hepatocytes. Intriguingly, Roc-A decreased LPS-induced production of reactive oxygen species (ROS), upregulated antioxidant gene expression, and downregulated endoplasmic reticulum (ER) stress-related gene expression. Mechanistically, Roc-A significantly attenuated LPS-induced phosphorylation of c-Jun N-terminal kinase (JNK) and activator protein-1 (AP-1). Notably, this effect was abolished by the JNK activator Anisomycin, while the JNK inhibitor SP600125 enhanced it. Furthermore, Roc-A suppressed the expression of NF-κB target genes, including inducible nitric oxide synthase (iNOS), thereby alleviating iNOS-derived nitric oxide (NO) production. These findings collectively indicate that Roc-A has the potential to alleviate LPS-induced nitrosative/oxidative stress and hepatic inflammation by inhibiting JNK phosphorylation. Thus, Roc-A emerges as a promising anti-inflammatory intervention for LPS-induced hepatic inflammation. Supplementary Information The online version contains supplementary material available at 10.1007/s43188-024-00263-y.
Collapse
Affiliation(s)
- Yoon-su Ha
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, 24341 Chuncheon, Republic of Korea
- Global/Gangwon Innovative Biologics-Regional Leading Research Center (GIB-RLRC), Kangwon National University, 24341 Chuncheon, Republic of Korea
| | - Taek-Kyong Kim
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, 24341 Chuncheon, Republic of Korea
- Global/Gangwon Innovative Biologics-Regional Leading Research Center (GIB-RLRC), Kangwon National University, 24341 Chuncheon, Republic of Korea
| | - Jun Heo
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, 24341 Chuncheon, Republic of Korea
- Global/Gangwon Innovative Biologics-Regional Leading Research Center (GIB-RLRC), Kangwon National University, 24341 Chuncheon, Republic of Korea
| | - Jintaek Oh
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, 24341 Chuncheon, Republic of Korea
- Global/Gangwon Innovative Biologics-Regional Leading Research Center (GIB-RLRC), Kangwon National University, 24341 Chuncheon, Republic of Korea
| | - Seung-Kyoon Kim
- Department of Convergent Bioscience and Informatics, College of Bioscience and Biotechnology, Chungnam National University, 34134 Daejeon, Republic of Korea
| | - Jeongkyu Kim
- Department of Life Science, Chung-Ang University, 06974 Seoul, Republic of Korea
| | - Jeonghyung Lee
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, 24341 Chuncheon, Republic of Korea
- Global/Gangwon Innovative Biologics-Regional Leading Research Center (GIB-RLRC), Kangwon National University, 24341 Chuncheon, Republic of Korea
| | - Se-Ran Yang
- Global/Gangwon Innovative Biologics-Regional Leading Research Center (GIB-RLRC), Kangwon National University, 24341 Chuncheon, Republic of Korea
- Department of Medicine, Kangwon National University, 24341 Chuncheon, Republic of Korea
| | - Seonghwan Hwang
- College of Pharmacy and Research Institute for Drug Development, Pusan National University, 46241 Busan, Republic of Korea
| | - Seung-Jin Kim
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, 24341 Chuncheon, Republic of Korea
- Global/Gangwon Innovative Biologics-Regional Leading Research Center (GIB-RLRC), Kangwon National University, 24341 Chuncheon, Republic of Korea
| |
Collapse
|
15
|
Jin S, Li Y, Xia T, Liu Y, Zhang S, Hu H, Chang Q, Yan M. Mechanisms and therapeutic implications of selective autophagy in nonalcoholic fatty liver disease. J Adv Res 2025; 67:317-329. [PMID: 38295876 PMCID: PMC11725165 DOI: 10.1016/j.jare.2024.01.027] [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: 12/03/2023] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 02/08/2024] Open
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) has become the most common chronic liver disease worldwide, whereas there is no approved drug therapy due to its complexity. Studies are emerging to discuss the role of selective autophagy in the pathogenesis of NAFLD, because the specificity among the features of selective autophagy makes it a crucial process in mitigating hepatocyte damage caused by aberrant accumulation of dysfunctional organelles, for which no other pathway can compensate. AIM OF REVIEW This review aims to summarize the types, functions, and dynamics of selective autophagy that are of particular importance in the initiation and progression of NAFLD. And on this basis, the review outlines the therapeutic strategies against NAFLD, in particular the medications and potential natural products that can modulate selective autophagy in the pathogenesis of this disease. KEY SCIENTIFIC CONCEPTS OF REVIEW The critical roles of lipophagy and mitophagy in the pathogenesis of NAFLD are well established, while reticulophagy and pexophagy are still being identified in this disease due to the insufficient understanding of their molecular details. As gradual blockage of autophagic flux reveals the complexity of NAFLD, studies unraveling the underlying mechanisms have made it possible to successfully treat NAFLD with multiple pharmacological compounds that target associated pathways. Overall, it is convinced that the continued research into selective autophagy occurring in NAFLD will further enhance the understanding of the pathogenesis and uncover novel therapeutic targets.
Collapse
Affiliation(s)
- Suwei Jin
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, China
| | - Yujia Li
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Tianji Xia
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, China
| | - Yongguang Liu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, China
| | - Shanshan Zhang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, China
| | - Hongbo Hu
- College of Food Science and Nutritional Engineering, China Agricultural University, China.
| | - Qi Chang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, China.
| | - Mingzhu Yan
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, China.
| |
Collapse
|
16
|
He Y, Xiao F, Yi B, Lu J. Prevalence and associated factors of MAFLD in adults with type 2 diabetes. PLOS GLOBAL PUBLIC HEALTH 2024; 4:e0003572. [PMID: 39775020 PMCID: PMC11684647 DOI: 10.1371/journal.pgph.0003572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 11/03/2024] [Indexed: 01/11/2025]
Abstract
To estimate the prevalence and associated factors of hepatic steatosis and fibrosis in adults with type 2 diabetes (T2DM) in the United States.Data were retrieved from the 2017‒March 2020 prepandemic cycle of the National Health and Nutritional Examination and Survey (NHANES). The study population included patients with T2DM. The controlled attenuation parameter (CAP) and liver stiffness measurement (LSM) were used to assess hepatic steatosis and fibrosis, respectively. A total of 1,290 T2DM patients were included, 85.2% (1044 patients) of whom presented with hepatic steatosis (CAP>248 dB/m). Among the 1044 T2DM patients with metabolically associated fatty liver disease (MAFLD), 29.5% developed hepatic fibrosis (LSM>8 kPa). Non-Hispanic black individuals (adjusted OR = 0.4008), BMI (adjusted OR = 1.1627), HbA1c (adjusted OR = 1.1450), TG (adjusted OR = 1.2347), HDL (adjusted OR = 0.4981), ALT (adjusted OR = 1.0227), AST (adjusted OR = 0.9396), and albumin (adjusted OR = 1.7030) were independently associated with steatosis. Age (adjusted OR = 1.0300), female sex (adjusted OR = 0.6655), BMI (adjusted OR = 1.1324), AST (adjusted OR = 1.0483), and GGT (adjusted OR = 1.0101) were independently associated with fibrosis. Heart failure was an independent factor associated with advanced fibrosis (adjusted OR = 1.9129) and cirrhosis (adjusted OR = 2.228). In the United States, hepatic steatosis is highly prevalent among T2DM patients, with 29.5% of these patients developing hepatic fibrosis. Some components of metabolic syndrome are related to hepatic steatosis and fibrosis. Moreover, heart failure is an independent factor associated with advanced fibrosis and cirrhosis.
Collapse
Affiliation(s)
- Yifei He
- Department of Endocrinology, Changhai Hospital, Naval Medical University, Shanghai, People’s Republic of China
| | - Feng Xiao
- Department of Liver Transplantation, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, People’s Republic of China
| | - Bin Yi
- Department of Liver Transplantation, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, People’s Republic of China
| | - Jin Lu
- Department of Endocrinology, Changhai Hospital, Naval Medical University, Shanghai, People’s Republic of China
| |
Collapse
|
17
|
Liu T, Sui M, Tian M, Wu N, Zhao S, Wang Y, Yang Y, Ma S, Jiao D, Wang L, Feng Y, Zhang Y, Qin C, Liu C, Qi J, Zhu Q. Sulfonated albumin from hepatocytes accelerates liver fibrosis in nonalcoholic fatty liver disease through endoplasmic reticulum stress. Free Radic Biol Med 2024; 228:150-162. [PMID: 39743026 DOI: 10.1016/j.freeradbiomed.2024.12.055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 12/15/2024] [Accepted: 12/28/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND Posttranslational modifications (PTM) of albumin occur in liver diseases; however, little is known about the source and function of sulfonated albumin, a significant modification of albumin occurring in nonalcoholic fatty liver disease (NAFLD). We aimed to investigate the mechanism underlying sulfonated albumin production and its role in the progression of NAFLD-related liver fibrosis. METHODS Serum samples from healthy controls and patients with NAFLD were used to measure the proportion of sulfonated albumin. Mice models with NAFLD fed with high-fat diet (HFD) and methionine choline-deficient diet (MCD) were constructed. RNA sequencing, KEGG analysis, and GSEA were used to explore the mechanism of sulfonated albumin production and its mechanism of activating hepatic stellate cells (HSCs) and promoting the progression of liver fibrosis in NAFLD. RESULTS Sulfonated albumin levels increased significantly in both human and mouse NAFLD serum samples. In vivo studies in mice have shown that the intraperitoneal injection of sulfonated albumin promotes inflammation, hepatic steatosis, and liver fibrosis in NAFLD. In addition, autophagy has been verified as a key mechanism in the regulation of sulfonated albumin production. We also demonstrated that reactive oxygen species (ROS) production depends on the accumulation of damaged mitochondria and affects the production of sulfonated albumin under the regulation of autophagy. Hepatocyte-derived sulfonated albumin activates HSCs through the GAL3 receptor, thereby activating the endoplasmic reticulum (ER) stress pathway and promoting profibrotic activation of HSCs. CONCLUSIONS Our study demonstrated that sulfonated albumin activated HSCs through GAL3, thereby accelerating NAFLD-related liver fibrosis. Serum sulfonated albumin may be a potential diagnostic marker for liver fibrosis and an important target for the treatment of NAFLD-related liver fibrosis.
Collapse
Affiliation(s)
- Tiantian Liu
- Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China; Department of Infectious Disease, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Minghao Sui
- Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China; Department of Infectious Disease, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China; Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Miaomiao Tian
- Department of Infectious Disease, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China; Department of Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Nijin Wu
- Department of Infectious Disease, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China; Department of Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Songbo Zhao
- Department of Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Yingchun Wang
- Department of Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Yinuo Yang
- Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China; Department of Infectious Disease, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China; Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Shujun Ma
- Department of Infectious Disease, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China; Department of Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China; Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Deyan Jiao
- Department of Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Le Wang
- Department of Health Care Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Yuemin Feng
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Yahui Zhang
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Chengyong Qin
- Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China; Department of Infectious Disease, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Chenxi Liu
- Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China; Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
| | - Jianni Qi
- Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China; Department of Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
| | - Qiang Zhu
- Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China; Department of Infectious Disease, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
| |
Collapse
|
18
|
Aydin I, Erisgin Z, Cinar E, Barak MZ, Tekelioglu Y, Usta M, Mutlu HS, Turkoglu I. Should combined MTX and CoQ10 use be reconsidered in terms of steatosis? A biochemical, flow cytometry, histopathological experimental study. Drug Chem Toxicol 2024:1-14. [PMID: 39734089 DOI: 10.1080/01480545.2024.2442660] [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: 10/21/2024] [Revised: 11/26/2024] [Accepted: 12/10/2024] [Indexed: 12/31/2024]
Abstract
In the present study, the effects of coenzyme Q10 (CoQ10), which is widely used in daily life, on the methotrexate (MTX)-induced hepatotoxicity, which is widely used today in malignancies and autoimmune diseases, were examined. Twenty-four female Wistar albino rats were divided into four groups. The group 1 (n = 6) was given 1 mL corn oil by oral gavage (p.o.) during seven days. Group 2 was given 20 mg/kg intraperitoneal (i.p.) MTX only on the first day of the experiment. Group 3 was given 20 mg/kg (i.p.) MTX on the first day of the experiment and 100 mg/kg CoQ10 dissolved in 1 mL corn oil were given by oral gavage during seven days, and group 4 was given 100 mg/kg CoQ10 dissolved in 1 mL corn oil by oral gavage during seven days. At the end of experiment, all animals were euthanized under anesthesia. In the liver tissue, histopathologic analysis on the hematoxylin and eosin (H&E), Masson trichrome, and periodic acid Schiff (PAS) stained sections, apoptotic analysis (% Annexin V positivity) by flow cytometry, and biochemical analysis for oxidative stress markers (GSH, CAT, and TBARS) was performed. According to histopathological analysis, apoptosis, concession, fibrosis, and inflammatory cell infiltration increased in the MTX group and those results significantly decreased in the MTX + CoQ10 groups. As an interesting result, fatty degeneration and TBARS elevation were observed in the MTX + CoQ10 group. As a result, although CoQ10 has protective effects on MTX-induced hepatotoxicity, fatty degeneration due to the combined usage of MTX and CoQ10 should be investigated with further studies.
Collapse
Affiliation(s)
- Ismail Aydin
- Department of General Surgery, Faculty of Medicine, Giresun University, Giresun, Turkiye
| | - Zuleyha Erisgin
- Department of Histology and Embryology, Faculty of Medicine, Giresun University, Giresun, Turkiye
| | - Esma Cinar
- Department of Pathology, Faculty of Medicine, Giresun University, Giresun, Turkiye
| | - M Zuhal Barak
- Department of Business, Adana Science and Technic University, Adana, Turkiye
| | - Yavuz Tekelioglu
- Department of Histology and Embryology, Faculty of Medicine, Black Sea Technical University, Trabzon, Turkiye
| | - Murat Usta
- Department of Medical Biochemistry, Faculty of Medicine, Giresun University, Giresun, Turkiye
| | - Hasan Serdar Mutlu
- Department of Histology and Embryology, Faculty of Medicine, Giresun University, Giresun, Turkiye
| | - Ismail Turkoglu
- Department of Histology and Embryology, Faculty of Medicine, Giresun University, Giresun, Turkiye
| |
Collapse
|
19
|
Tong W, Zhu L, Han P, Bai Y, Wang T, Chen D, Li Z, Chi H, Deng X, Zhang Y, Shen Z. TWEAK is an activator of Hippo-YAP signaling protecting against hepatic Ischemia/ reperfusion injury. Int Immunopharmacol 2024; 143:113567. [PMID: 39500083 DOI: 10.1016/j.intimp.2024.113567] [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: 07/31/2024] [Revised: 10/11/2024] [Accepted: 10/31/2024] [Indexed: 12/08/2024]
Abstract
Hepatic ischemia-reperfusion injury (IRI) represents a formidable complication commonly linked with hemorrhagic shock, liver resection, and transplantation. This study aims to elucidate the role of Tumor Necrosis Factor-like Weak Inducer of Apoptosis (TWEAK) in the pathogenesis of hepatic I/R injury and to delineate the underlying mechanisms involved. Utilizing a hypoxia-reoxygenation model in human liver organoids (HLOs) alongside a murine model of warm ischemia-reperfusion injury, we systematically investigated the interplay between TWEAK, its receptor Fn14, and the HIPPO signaling pathway. Our findings indicate that TWEAK pretreatment significantly mitigates IRI in murine livers as well as hypoxia/reoxygenation injury in HLOs. Notably, administration of adeno-associated virus (AAV) to knock down Fn14 abrogated the protective effects of TWEAK in the murine model. Transcriptome sequencing analysis revealed that the interaction between TWEAK and Fn14 enhances cellular resistance to IRI by activating the HIPPO signaling pathway. Overall, TWEAK emerges as a promising therapeutic target for mitigating hepatic I/R injury, potentially improving outcomes in liver transplantation.
Collapse
Affiliation(s)
- Wen Tong
- The First Central Clinical School, Tianjin Medical University, Tianjin 300070, China
| | - Liuyang Zhu
- The First Central Clinical School, Tianjin Medical University, Tianjin 300070, China
| | - Pinsheng Han
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Yi Bai
- Department of Hepatobiliary Surgery, Tianjin First Central Hospital, Tianjin 300192, China
| | - Tianze Wang
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Dapeng Chen
- The First Central Clinical School, Tianjin Medical University, Tianjin 300070, China
| | - Zhongmin Li
- Department of Hepatobiliary Surgery, Tianjin Nankai Hospital, Tianjin 300100, China
| | - Hao Chi
- The First Central Clinical School, Tianjin Medical University, Tianjin 300070, China
| | - Xiyue Deng
- The First Central Clinical School, Tianjin Medical University, Tianjin 300070, China
| | - Yamin Zhang
- Department of Hepatobiliary Surgery, Tianjin First Central Hospital, Tianjin 300192, China.
| | - Zhongyang Shen
- Organ Transplantation Centre, Tianjin First Central Hospital, Tianjin 300192, China.
| |
Collapse
|
20
|
Wang C, Feng X, Li W, Chen L, Wang X, Lan Y, Tang R, Jiang T, Zheng L, Liu G. Apigenin as an emerging hepatoprotective agent: current status and future perspectives. Front Pharmacol 2024; 15:1508060. [PMID: 39749193 PMCID: PMC11693974 DOI: 10.3389/fphar.2024.1508060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 12/04/2024] [Indexed: 01/04/2025] Open
Abstract
Apigenin (C15H10O5, API) is a natural flavonoid widely found in vegetables, fruits, and plants such as celery, oranges, and chamomile. In recent years, API has attracted considerable attention as a dietary supplement due to its low toxicity, non-mutagenic properties and remarkable therapeutic efficacy in various diseases. In particular, evidence from a large number of preclinical studies suggests that API has promising effects in the prevention and treatment of a variety of liver diseases, including multifactorial liver injury, non-alcoholic fatty liver disease/non-alcoholic steatohepatitis, liver fibrosis and liver cancer. This paper provides a comprehensive review of the progress of research into the therapeutic applications of API in liver diseases as of August 2024, based on literature retrieved from databases such as Web of Science, PubMed, CNKI, Google Scholar and ScienceDirect. The hepatoprotective effects of API involve multiple molecular mechanisms, including inhibition of inflammation, alleviation of hepatic oxidative stress, amelioration of insulin resistance, promotion of fatty acid oxidation, inhibition of liver cancer cell proliferation and differentiation, and induction of tumour cell apoptosis. More importantly, signaling pathways such as Nrf2, NF-κB, PI3K/Akt/mTOR, NLRP3, Wnt/β-catenin, TGF-β1/Smad3, AMPK/SREBP, PPARα/γ, MAPKs, and Caspases are identified as key targets through which API exerts its beneficial effects in various liver diseases. Studies on its toxicity and pharmacokinetics indicate that API has low toxicity, is slowly metabolized and excreted in vivo, and has low oral bioavailability. In addition, the paper summarises and discusses the sources, physicochemical properties, new dosage forms, and current challenges and opportunities of API, with the aim of providing direction and rationale for the further development and clinical application of API in the food, pharmaceutical and nutraceutical fields.
Collapse
Affiliation(s)
- Cheng Wang
- School of Clinical Medical, Chengdu Medical College, Chengdu, China
- Department of Pharmacy, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Xiaoli Feng
- School of Clinical Medical, Chengdu Medical College, Chengdu, China
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Wen Li
- School of Clinical Medical, Chengdu Medical College, Chengdu, China
- Department of Pharmacy, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Li Chen
- School of Clinical Medical, Chengdu Medical College, Chengdu, China
- Department of Pharmacy, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Xinming Wang
- School of Clinical Medical, Chengdu Medical College, Chengdu, China
- Department of Pharmacy, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Yimiao Lan
- School of Clinical Medical, Chengdu Medical College, Chengdu, China
- Department of Pharmacy, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Rong Tang
- College of Foreign Languages and Cultures, Sichuan University, Chengdu, China
| | - Ting Jiang
- School of Clinical Medical, Chengdu Medical College, Chengdu, China
- Department of Pharmacy, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Lingli Zheng
- School of Clinical Medical, Chengdu Medical College, Chengdu, China
- Department of Pharmacy, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Gang Liu
- School of Clinical Medical, Chengdu Medical College, Chengdu, China
- Department of Pharmacy, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| |
Collapse
|
21
|
Bucci T, Nabrdalik K, Baratta F, Pastori D, Pignatelli P, Hydes T, Alam U, Violi F, Lip GYH. Risk of Adverse Events in Anticoagulated Patients With Atrial Fibrillation and Nonalcoholic Fatty Liver Disease. J Clin Endocrinol Metab 2024; 110:208-217. [PMID: 38864452 PMCID: PMC11651694 DOI: 10.1210/clinem/dgae394] [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: 03/12/2024] [Revised: 05/15/2024] [Accepted: 06/07/2024] [Indexed: 06/13/2024]
Abstract
BACKGROUND The clinical impact of nonalcoholic fatty liver disease (NAFLD) in patients with atrial fibrillation (AF) is still controversial. AIM To evaluate the 1-year risk of all-cause death, thromboembolic events, and bleeding in patients with AF-NAFLD. METHODS Retrospective study with a health research network (TriNetX). Patients with AF on oral anticoagulation (OAC) were categorized according to the presence of NAFLD into 2 groups. The primary outcomes were the 1-year risks of (1) a composite cardiovascular outcome (all-cause death, myocardial infarction, stroke, cardiac arrest, and pulmonary embolism) and (2) a composite hemorrhagic outcome (intracranial hemorrhage and gastrointestinal bleeding). Cox regression analysis before and after propensity score matching was used to estimate hazard ratio (HR) and 95% 95% CI,. Sensitivity analyses investigated the risk associated with cirrhosis, thrombocytopenia, and type of OAC (warfarin vs non-vitamin K antagonist oral anticoagulants (NOACs). RESULTS We identified 22 636 patients with AF-NAFLD (69 ± 12 years, 46.7% females) and 391 014 patients with AF and without liver disease (72 ± 12 years, 42.7% females). NAFLD was associated with a higher risk of composite cardiovascular (HR, 1.54; 95% CI, 1.47-1.61) and hemorrhagic (HR, 1.56; 95% CI, 1.42-1.72) outcomes. This was consistent also for all the single outcomes. Cirrhotic and thrombocytopenic patients with AF-NAFLD showed the highest risks. Compared to patients with AF-NAFLD on NOACs, those on warfarin were associated with a higher risk of cardiovascular and hemorrhagic outcomes. CONCLUSION In patients with AF, NAFLD is associated with a higher 1-year risk of adverse events, with the risk of adverse events progressively increasing from noncirrhotic to cirrhotic and from nonthrombocytopenic to thrombocytopenic patients. NOACs were associated with a better effectiveness and safety profile compared to warfarin.
Collapse
Affiliation(s)
- Tommaso Bucci
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool and Heart and Chest Hospital, Liverpool L7 8TX, UK
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome 00185, Italy
| | - Katarzyna Nabrdalik
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool and Heart and Chest Hospital, Liverpool L7 8TX, UK
- Department of Internal Medicine, Diabetology and Nephrology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Katowice 40-055, Poland
| | - Francesco Baratta
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome 00185, Italy
| | - Daniele Pastori
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome 00185, Italy
| | - Pasquale Pignatelli
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome 00185, Italy
| | - Theresa Hydes
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool and Heart and Chest Hospital, Liverpool L7 8TX, UK
- Diabetes & Endocrinology Research and Pain Research Institute, Institute of Life Course and Medical Sciences, University of Liverpool and Liverpool University Hospital NHS Foundation Trust, Liverpool L69 7ZX, UK
| | - Uazman Alam
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool and Heart and Chest Hospital, Liverpool L7 8TX, UK
- Diabetes & Endocrinology Research and Pain Research Institute, Institute of Life Course and Medical Sciences, University of Liverpool and Liverpool University Hospital NHS Foundation Trust, Liverpool L69 7ZX, UK
| | - Francesco Violi
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome 00185, Italy
| | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool and Heart and Chest Hospital, Liverpool L7 8TX, UK
- Danish Center for Health Services Research, Department of Clinical Medicine, Aalborg University, Aalborg 9000, Denmark
| |
Collapse
|
22
|
Mu C, Wang S, Wang Z, Tan J, Yin H, Wang Y, Dai Z, Ding D, Yang F. Mechanisms and therapeutic targets of mitochondria in the progression of metabolic dysfunction-associated steatotic liver disease. Ann Hepatol 2024:101774. [PMID: 39701281 DOI: 10.1016/j.aohep.2024.101774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 12/13/2024] [Accepted: 12/14/2024] [Indexed: 12/21/2024]
Abstract
Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD) includes liver disease processes from simple fatty liver to nonalcoholic steatohepatitis, which may progress to liver fibrosis, cirrhosis, and even hepatocellular carcinoma (HCC). As the incidence of HCC derived from viral hepatitis decreases, MASLD has emerged as a significant health threat, driven by lifestyle changes and rising obesity rates among patients. The pathogenesis of MASLD is complex, involving factors such as insulin resistance, gut microbiota imbalance, and genetic and epigenetic factors. In recent years, the role of mitochondrial dysfunction in MASLD has gained significant attention, involving β-oxidation imbalance, oxidative stress increase, mitophagy defects, and mitochondrial DNA (mtDNA) mutations. This article reviews the pathophysiological mechanisms of mitochondrial dysfunction in MASLD, diagnostic methods, and potential therapeutic strategies. By synthesizing current research findings, the review aims to highlight the critical role of mitochondrial dysfunction as a target for future diagnostic and therapeutic interventions. This focus could pave the way for innovative clinical strategies, ultimately improving treatment options and patient prognosis in MASLD.
Collapse
Affiliation(s)
- Chenyang Mu
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China; Department of Medical Genetics, Naval Medical University, Shanghai, China
| | - Sijie Wang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China; Department of Medical Genetics, Naval Medical University, Shanghai, China
| | - Zenghan Wang
- Department of Medical Genetics, Naval Medical University, Shanghai, China
| | - Jian Tan
- Department of Medical Genetics, Naval Medical University, Shanghai, China
| | - Haozan Yin
- Department of Medical Genetics, Naval Medical University, Shanghai, China
| | - Yuefan Wang
- Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Zhihui Dai
- Department of Medical Genetics, Naval Medical University, Shanghai, China
| | - Dongyang Ding
- Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Fu Yang
- Department of Medical Genetics, Naval Medical University, Shanghai, China; Shanghai Key Laboratory of Medical Bioprotection, Shanghai, China; Key Laboratory of Biological Defense, Ministry of Education, Shanghai, China.
| |
Collapse
|
23
|
Niu QQ, Xi YT, Zhang CR, Li XY, Li CZ, Wang HD, Li P, Yin YL. Potential mechanism of perillaldehyde in the treatment of nonalcoholic fatty liver disease based on network pharmacology and molecular docking. Eur J Pharmacol 2024; 985:177092. [PMID: 39510336 DOI: 10.1016/j.ejphar.2024.177092] [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: 05/30/2024] [Revised: 10/08/2024] [Accepted: 10/31/2024] [Indexed: 11/15/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is one of the most common chronic metabolic liver diseases worldwide. Perillaldehyde (4-propyl-1-en-2-ylcyclohexene-1-aldehyde, PA) is a terpenoid compound extracted from Perilla, which has effective pharmacological activities such as anti-inflammatory, antidepressant, and anticancer. This study aimed to explore the pharmacological effects of PA in intervening with NAFLD and reveal its potential mechanisms. Firstly, we identified the core targets of PA intervention therapy for NAFLD through network pharmacology and molecular docking techniques. After that, in vitro animal experiments such as H&E and Masson staining, immunofluorescence, immunohistochemistry, and Western blot were conducted to validate the results network effectively pharmacology predicted. Network pharmacology analysis suggested that PPAR-α may be the core target of PA intervention in NAFLD. H&E and Masson staining showed that after low-dose (50 mg/kg) PA administration, there was a noticeable improvement in fat deposition in the livers of NAFLD mice, and liver tissue fibrosis was alleviated. Immunohistochemical and immunofluorescence analysis showed that low dose (50 mg/kg) PA could reduce hepatocyte apoptosis, decrease the content of pro-apoptosis protein Bax, and increase the expression of anti-apoptosis protein Bcl-2 in NAFLD mice. Western blot results confirmed that low-dose (50 mg/kg) PA could increase the expression of PPAR-α and inhibit the expression of NF-κB in NAFLD mice. Our study indicated that PA could enhance the activity of PPAR-α and reduce the level of NF-κB in NAFLD mice, which may positively affect the prevention of NAFLD.
Collapse
Affiliation(s)
- Qian-Qian Niu
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China; Department of Toxicology, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Penang, 13200, Malaysia
| | - Yu-Ting Xi
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, 453003, China
| | - Chun-Rui Zhang
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, 453003, China
| | - Xi-Yue Li
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, 453003, China
| | - Cheng-Zhi Li
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, 453003, China
| | - Hui-Dan Wang
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, 453003, China
| | - Peng Li
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, 453003, China.
| | - Ya-Ling Yin
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China.
| |
Collapse
|
24
|
Mignini I, Galasso L, Piccirilli G, Calvez V, Termite F, Esposto G, Borriello R, Miele L, Ainora ME, Gasbarrini A, Zocco MA. Interplay of Oxidative Stress, Gut Microbiota, and Nicotine in Metabolic-Associated Steatotic Liver Disease (MASLD). Antioxidants (Basel) 2024; 13:1532. [PMID: 39765860 PMCID: PMC11727446 DOI: 10.3390/antiox13121532] [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: 09/29/2024] [Revised: 12/10/2024] [Accepted: 12/12/2024] [Indexed: 01/15/2025] Open
Abstract
Oxidative stress has been described as one of the main drivers of intracellular damage and metabolic disorders leading to metabolic syndrome, a major health problem worldwide. In particular, free radicals alter lipid metabolism and promote lipid accumulation in the liver, existing in the hepatic facet of metabolic syndrome, the metabolic dysfunction-associated steatotic liver disease (MASLD). Recent literature has highlighted how nicotine, especially if associated with a high-fat diet, exerts a negative effect on the induction and progression of MASLD by upregulating inflammation and increasing oxidative stress, abdominal fat lipolysis, and hepatic lipogenesis. Moreover, considerable evidence shows the central role of intestinal dysbiosis in the pathogenesis of MASLD and the impact of nicotine-induced oxidative stress on the gut microbiome. This results in an intricate network in which oxidative stress stands at the intersection point between gut microbiome, nicotine, and MASLD. The aim of this review is to delve into the molecular mechanisms linking tobacco smoking and MASLD, focusing on nicotine-induced microbiota modifications and their impact on MASLD development.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Maria Assunta Zocco
- CEMAD Digestive Diseases Center, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168 Rome, Italy; (I.M.); (L.G.); (G.P.); (V.C.); (F.T.); (G.E.); (R.B.); (L.M.); (M.E.A.); (A.G.)
| |
Collapse
|
25
|
Zhang SH, Zhang HJ, Jia YZ, Wang ZY, You ZH, Lian CY, Wang L. Melatonin prevents glyphosate-induced hepatic lipid accumulation in roosters via activating Nrf2 pathway. Int Immunopharmacol 2024; 142:113180. [PMID: 39305889 DOI: 10.1016/j.intimp.2024.113180] [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: 06/14/2024] [Revised: 08/28/2024] [Accepted: 09/12/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND Glyphosate (GLY) is a widely used herbicide with well-defined hepatotoxic effects, in which oxidative stress has been shown to be involved in the pathogenesis of hepatotoxicity. Melatonin (MET), an effective free radical scavenger, has been revealed to alleviate drug-induced liver damage by inhibiting oxidative stress. METHODS In this study, a rooster model with primary chicken embryo hepatocytes was applied to elucidate the therapeutic effects of MET against GLY-induced hepatic damage and the potential mechanism. Histopathological examinations, biochemical tests and immunoblotting analysis were used to monitor the protective effects of MET on GLY-induced hepatic lipid accumulation. Molecular docking analysis was used to reveal the key reason of MET-improved hepatic lipid deposition. RESULTS Data firstly showed that MET administration markedly improved GLY-induced hepatic injury, as evidenced by normalized liver enzymes and alleviated pathological changes of liver tissues. Moreover, MET supplementation alleviated GLY-induced hepatic lipid accumulation, which was correlated with improved serum and hepatic lipid profiles and normalized expression of lipolysis- and lipogenesis-related proteins. Notably, MET significantly inhibited vital enzymes involved in stimulating oxidative stress. Moreover, MET enhanced GLY-inhibited Nrf2 nuclear transcription and increased the expressions of its downstream target genes HO1 and NQO1. Further studies revealed that MET may interact with Nrf2 to enhance nuclear translocation of Nrf2. CONCLUSION Collectively, our results provide the first direct evidence that MET is a novel regulator of Nrf2, highlighting that Nrf2 may be a potential therapeutic target for GLY-induced lipotoxic liver injury.
Collapse
Affiliation(s)
- Shu-Hui Zhang
- College of Veterinary Medicine, Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, 7 Panhe Street, Tai'an City, Shandong Province 271017, China
| | - Hai-Jing Zhang
- New Drug Evaluation Center of Shandong Academy of Pharmaceutical Sciences, Shandong Academy of Pharmaceutical Sciences, 989 Xinluo Street, Ji'nan City 250101, Shandong Province, China
| | - Yan-Zhan Jia
- College of Veterinary Medicine, Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, 7 Panhe Street, Tai'an City, Shandong Province 271017, China
| | - Zhen-Yong Wang
- College of Veterinary Medicine, Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, 7 Panhe Street, Tai'an City, Shandong Province 271017, China
| | - Zhao-Hong You
- Zaozhuang University School of Food Science and Pharmaceutical Engineering, No.1, Beian Road, Shizhong District, Zaozhuang City, Shandong Province 277160, China
| | - Cai-Yu Lian
- College of Veterinary Medicine, Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, 7 Panhe Street, Tai'an City, Shandong Province 271017, China.
| | - Lin Wang
- College of Veterinary Medicine, Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, 7 Panhe Street, Tai'an City, Shandong Province 271017, China.
| |
Collapse
|
26
|
Zhu J, Liao X, Du L, Lv P, Deng J. Associations of serum folate and vitamin B 12 levels with all-cause mortality among patients with metabolic dysfunction associated steatotic liver disease: a prospective cohort study. Front Endocrinol (Lausanne) 2024; 15:1426103. [PMID: 39703860 PMCID: PMC11655224 DOI: 10.3389/fendo.2024.1426103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 11/11/2024] [Indexed: 12/21/2024] Open
Abstract
Introduction Serum folate and vitamin B12 levels correlate with the prevalence of fatty liver disease, but it is not clear how they affect mortality. Therefore, this study aimed to investigate the association of serum folate and vitamin B12 concentrations with all-cause mortality in individuals with metabolic dysfunction-associated steatotic liver disease (MASLD). Methods MASLD subjects were from the Third National Health and Nutrition Examination Survey (NHANES III) in the United States, and mortality follow-up data were obtained by linkage to death records from the National Death Index. Multivariable Cox proportional regression models and restricted cubic spline (RCS) models were used to evaluate the association of serum folate/vitamin B12 with all-cause mortality in the MASLD population. Results 3,636 and 2,125 MASLD individuals were included in the analyses related to serum folate and vitamin B12, respectively. During a follow-up period of more than 20 years, the RCS models demonstrated significant nonlinear associations of both serum folate (P <0.001) and vitamin B12 (P =0.016) with all-cause mortality in MASLD. When their serum concentrations were below the median level, the risk of all-cause mortality decreased with increasing concentration, reaching a lowest risk around the median level, and then leveled off. In the multivariable cox regression model, for vitamin B12, the risk of all-cause mortality was reduced by 42% and 28% in the third and fourth quartile groups, respectively, compared with the lowest quartile group (hazard ratio [HR]=0.58, 95% CI: 0.39-0.86, P =0.008; HR =0.72, 95% CI: 0.54-0.96, P=0.026, respectively). For folate, the risk of all-cause mortality was reduced by 28% in the third quartile compared with the lowest quartile (HR =0.72, 95% CI: 0.57-0.91, P =0.005). Conclusion This longitudinal cohort study suggests that low serum folate and vitamin B12 levels in patients with MASLD are significantly associated with an elevated risk of all-cause mortality.
Collapse
Affiliation(s)
- Jiaxin Zhu
- Department of Clinical Laboratory, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Xinyi Liao
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Lei Du
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Pengju Lv
- Department of Clinical Laboratory, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Jian Deng
- Department of Thyroid Breast Surgery, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
27
|
Zhang R, Sun J, Wang Y, Yu H, Wang S, Feng X. Ameliorative effect of phenolic compound-pterostilbene on corticosterone-induced hepatic lipid metabolic disorder in broilers. J Nutr Biochem 2024; 137:109822. [PMID: 39645170 DOI: 10.1016/j.jnutbio.2024.109822] [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: 08/09/2024] [Revised: 10/28/2024] [Accepted: 12/01/2024] [Indexed: 12/09/2024]
Abstract
The aim of this study was to investigate the ameliorative effects of pterostilbene (PTE), a polyphenolic compound, on stress-induced lipid metabolic disorders in the liver of broiler chickens. Six hundred healthy, 1-day-old Arbor Acres with similar weight were randomly assigned to five groups, each consisting of eight replicates with 15 broilers per replicate. The groups included: a control group (fed a basal diet), and four groups treated with corticosterone (CORT) at varying dietary levels of PTE supplementation: CORT (0 mg/kg PTE), CORT-PT200 (200 mg/kg PTE), CORT-PT400 (400 mg/kg PTE), and CORT-PT600 (600 mg/kg PTE). The results indicated that PTE administration to corticosterone (CORT)-injected broilers significantly improved weight gain, reduced liver index, and lowered the elevation of serum aspartate aminotransferase, gamma-glutamyl transferase, glucose, total cholesterol, triglycerides, and lipoprotein cholesterol concentrations induced by CORT injection (P<.05), but had no significant effect on serum CORT concentration (P>.05). PTE also significantly reduced the increased rate of abdominal fat deposition induced by CORT, decreased the average size of adipocytes, and downregulated the expression of the FAS gene (P<.05). It reversed the increase in liver total cholesterol, triglycerides, lipoprotein cholesterol, and non-esterified fatty acids content induced by CORT (P<.05). PTE had no significant effect on the expression of the glucocorticoid receptor (P>.05), but significantly upregulated the protein expression of Sirt1 and p-AMPK (P<.05), promoted the expression of lipid autophagy genes MAP1LC3B and lipolytic genes LPL, but inhibited the expression of fatty acid synthesis genes SREBP-1c, ACC, and SCD (P<.05). The addition of PTE to the diet alleviated CORT-induced oxidative stress and inflammation by enhancing T-SOD and GSH-Px activities, reducing MDA content, inhibiting p-NF-κB p65 and NLRP3 expression and the release of TNF-α and IL-1β in the serum, and increasing IL-4 content (P<.05). Overall, dietary PTE effectively regulates lipid metabolism and antioxidant status, offering a potential strategy to mitigate stress-induced metabolic disruptions in broilers.
Collapse
Affiliation(s)
- Ruoshi Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Jing Sun
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Yingjie Wang
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Hao Yu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Shenao Wang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Xingjun Feng
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China.
| |
Collapse
|
28
|
Peng H, Zhao M, Zhang Y, Guo Y, Zhao A. Increased magnesium intake does not mitigate MAFLD risk associated with magnesium deficiency. Sci Rep 2024; 14:30386. [PMID: 39639030 PMCID: PMC11621767 DOI: 10.1038/s41598-024-82203-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 12/03/2024] [Indexed: 12/07/2024] Open
Abstract
Serum magnesium cannot accurately assess magnesium deficiency. The association between magnesium deficiency and metabolic dysfunction-associated fatty liver disease (MAFLD) remains unclear. Selecting 3,377 participants in the United States, we assessed the degree of magnesium deficiency in the population using magnesium depletion score (MDS). Multinomial logistic regression assessed the association between magnesium deficiency and MAFLD. Subgroup analyses assessed the association between dietary magnesium intake and MAFLD under different magnesium deficiency statuses. Structural equation modeling (SEM) revealed mediation effects. Magnesium deficiency was associated with MAFLD (ORseverevs.none: 1.69, 95%CI: 1.16-2.46; p for trend < 0.001). Magnesium intake was negatively associated with MAFLD only in the subgroup without magnesium deficiency (p for trend < 0.01). Inflammation, oxidative stress, and aging significantly mediated the association between MDS and MAFLD (all p < 0.05). In American adults, magnesium deficiency assessed by MDS might be a risk factor for MAFLD, with inflammation, oxidative stress, and aging potentially being key mechanisms. Simply increasing magnesium intake would not mitigate MAFLD risk associated with magnesium deficiency. Correcting magnesium deficiency might prevent MAFLD.
Collapse
Affiliation(s)
- Haiyang Peng
- Department of Hepatobiliary Surgery, Second Hospital Affiliated to Chongqing Medical University, Chongqing, P. R. China
- Kuanren Laboratory of Translational lipidology, Centre for Lipid Research, Second Hospital, Chongqing Medical University, Chongqing, P. R. China
| | - Minjie Zhao
- Department of Hepatobiliary Surgery, Second Hospital Affiliated to Chongqing Medical University, Chongqing, P. R. China
- Kuanren Laboratory of Translational lipidology, Centre for Lipid Research, Second Hospital, Chongqing Medical University, Chongqing, P. R. China
| | - Yuezhou Zhang
- Department of Hepatobiliary Surgery, Second Hospital Affiliated to Chongqing Medical University, Chongqing, P. R. China
- Kuanren Laboratory of Translational lipidology, Centre for Lipid Research, Second Hospital, Chongqing Medical University, Chongqing, P. R. China
| | - Yaoyuan Guo
- Department of Clinical Nutrition, The First Affiliated Hospital of Chongqing Medical and Pharmaceutical College, Chongqing, P. R. China
| | - Andong Zhao
- Kuanren Laboratory of Translational lipidology, Centre for Lipid Research, Second Hospital, Chongqing Medical University, Chongqing, P. R. China.
- Department of Plastic and Maxillofacial Surgery, Second Hospital Affiliated to Chongqing Medical University, Chongqing, P. R. China.
| |
Collapse
|
29
|
Hur YK, Lee HE, Yoo JY, Park YN, Lee IH, Bae YS. NADPH oxidase 4-SH3 domain-containing YSC84-like 1 complex participates liver inflammation and fibrosis. Free Radic Biol Med 2024; 227:246-259. [PMID: 39645205 DOI: 10.1016/j.freeradbiomed.2024.12.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 11/19/2024] [Accepted: 12/04/2024] [Indexed: 12/09/2024]
Abstract
There is growing evidence that NADPH oxidase 4 (Nox4) in hepatocytes contributes to liver inflammation and fibrosis during the development of metabolic dysfunction-associated steatohepatitis (MASH). However, how Nox4 is regulated and leads to liver pathogenesis is unclear. Our previous studies showed that the cytosolic protein SH3 domain-containing Ysc84-like 1 (SH3YL1) regulates Nox4 activity. Here, we asked whether SH3YL1 also participates in liver inflammation and fibrosis during MASH development. We generated that whole body SH3YL1 knockout (SH3YL1-/-), Nox4 knockout (Nox4-/-) mice, and the hepatocyte-specific SH3YL1 conditional knockout (Alb-Cre/SH3YL1fl/fl) mice were fed a methionine/choline-deficient (MCD) diet to induce liver inflammation and fibrosis in pathogenesis of MASH. Palmitate-stimulated primary SH3YL1-and Nox4-deficient hepatocytes and hepatic stellate cells (HSCs) did not generate H2O2. While the liver of MCD diet-fed wild type (WT) mice demonstrated elevated 3-nitrotyrosine as a protein oxidation and 4-hydroxynonenal adducts as a lipid oxidation and increased liver inflammation, hepatocyte apoptosis, and liver fibrosis, these events were markedly reduced in SH3YL1-/-, Nox4-/-, and Alb-Cre/SH3YL1fl/fl mice. The MCD diet-fed WT mice also showed elevated hepatocyte expression of SH3YL1 protein. Similarly, liver biopsies from MASH patients demonstrated strong hepatocyte SH3YL1 protein expression, whereas hepatocytes from patients with steatosis weakly expressed SH3YL1 and histologically normal patient hepatocytes exhibited very little SH3YL1 expression. The Nox4-SH3YL1 complex in murine hepatocytes elevates their H2O2 production, which promotes the liver inflammation, hepatocyte apoptosis, and liver fibrosis that characterize MASH. This axis may also participate in MASH in humans.
Collapse
Affiliation(s)
- Yeo Kyu Hur
- Department of Life Sciences, Ewha Womans University, 52 Ewhayeodae-Gil, Seodaemoon-Gu, Seoul, 03760, South Korea
| | - Hye Eun Lee
- Celros Biotech, 52 Ewhayeodae-Gil, Seodaemoon-Gu, Seoul, 03760, South Korea
| | - Jung-Yeon Yoo
- Department of Life Sciences, Ewha Womans University, 52 Ewhayeodae-Gil, Seodaemoon-Gu, Seoul, 03760, South Korea
| | - Young Nyun Park
- Department of Pathology Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-Gu, Seoul, 03722, South Korea
| | - In Hye Lee
- Department of Life Sciences, Ewha Womans University, 52 Ewhayeodae-Gil, Seodaemoon-Gu, Seoul, 03760, South Korea.
| | - Yun Soo Bae
- Department of Life Sciences, Ewha Womans University, 52 Ewhayeodae-Gil, Seodaemoon-Gu, Seoul, 03760, South Korea; Celros Biotech, 52 Ewhayeodae-Gil, Seodaemoon-Gu, Seoul, 03760, South Korea.
| |
Collapse
|
30
|
Mouskeftara T, Kalopitas G, Liapikos T, Arvanitakis K, Theocharidou E, Germanidis G, Gika H. A Comprehensive Analysis of Liver Lipidomics Signature in Adults with Metabolic Dysfunction-Associated Steatohepatitis-A Pilot Study. Int J Mol Sci 2024; 25:13067. [PMID: 39684777 DOI: 10.3390/ijms252313067] [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: 10/20/2024] [Revised: 12/01/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD) is the most common chronic liver disorder in Western countries, encompassing a range of conditions from steatosis to Metabolic Dysfunction-Associated Steatohepatitis (MASH), which can potentially progress to cirrhosis. Lipidomics approaches have revealed significant alterations in the hepatic lipidome associated with both steatosis and steatohepatitis, with these changes correlating with disease manifestation. While the transition from steatosis to MASH remains poorly understood, recent research indicates that both the quantity and quality of deposited lipids play a pivotal role in MASLD progression. In our study, we utilized untargeted and targeted analyses to identify intact lipids and fatty acids in liver biopsies from healthy controls and MASLD patients, categorized based on their histological findings. In total, 447 lipid species were identified, with 215 subjected to further statistical analysis. Univariate and multivariate analyses revealed alterations in triglyceride species and fatty acids, including FA 16:0, FA 16:1, FA 18:3 n6, the sum of MUFA, and the Δ9-desaturase activity ratio. This research provides insights into the connection between dysregulated lipid metabolism in the progression of MASLD, supporting previous findings. Further studies on lipid metabolism could improve risk assessment methods, particularly given the current limited understanding of the transition from steatosis to MASH.
Collapse
Affiliation(s)
- Thomai Mouskeftara
- Laboratory of Forensic Medicine & Toxicology, Department of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Georgios Kalopitas
- Division of Gastroenterology and Hepatology, 1st Department of Internal Medicine, AHEPA University Hospital, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
- Basic and Translational Research Unit, Special Unit for Biomedical Research and Education, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
- Laboratory of Hygiene, Social and Preventive Medicine and Medical Statistics, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Theodoros Liapikos
- Laboratory of Analytical Chemistry, Department of Chemistry, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Konstantinos Arvanitakis
- Division of Gastroenterology and Hepatology, 1st Department of Internal Medicine, AHEPA University Hospital, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
- Basic and Translational Research Unit, Special Unit for Biomedical Research and Education, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Eleni Theocharidou
- 2nd Department of Internal Medicine, Hippokration General Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
| | - Georgios Germanidis
- Division of Gastroenterology and Hepatology, 1st Department of Internal Medicine, AHEPA University Hospital, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
- Basic and Translational Research Unit, Special Unit for Biomedical Research and Education, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
- Laboratory of Hygiene, Social and Preventive Medicine and Medical Statistics, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Helen Gika
- Laboratory of Forensic Medicine & Toxicology, Department of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
- Biomic AUTh, Center for Interdisciplinary Research and Innovation (CIRI-AUTH), Balkan Center B1.4, 10th km Thessaloniki-Thermi Rd., 57001 Thessaloniki, Greece
| |
Collapse
|
31
|
Wei Y, Pan T, Zhao Y, Chen Z, Wu L, Fang S, Wang X, Wang X, Chen D, Chen Y. Nicotine aggravates high-fat diet-induced non-alcoholic fatty liver disease in mice via inhibition of CISD3. Int Immunopharmacol 2024; 142:113067. [PMID: 39241515 DOI: 10.1016/j.intimp.2024.113067] [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: 05/08/2024] [Revised: 08/01/2024] [Accepted: 08/30/2024] [Indexed: 09/09/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most prevalent chronic liver disease globally. Growing data suggests that smoking plays an important role in the evolution of NAFLD. CDGSH iron sulfur domain 3 (CISD3) regulates critical biological activities. However, its role in nicotine-associated NAFLD and its underlying mechanisms have not been elucidated. Mice were given a high-fat diet for 10 weeks to induce the development of NAFLD. The results revealed that in mice with NAFLD, nicotine treatment resulted in reduced CISD3 expression, leading to mitochondrial dysfunction and impaired β-oxidation. Notably, exacerbation of hepatic steatosis and inflammatory injury was observed. Furthermore, Cisd3-knockout exacerbated lipid accumulation, aggravating oxidative stress and apoptosis. In conclusion, these results contribute to our knowledge of the function of CISD3 in nicotine-associated NAFLD, revealing the possibility of using CISD3 as a potential molecular target for treating NAFLD.
Collapse
Affiliation(s)
- Yifeng Wei
- Hepatology Diagnosis and Treatment Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou 325035, China
| | - Tongtong Pan
- Hepatology Diagnosis and Treatment Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou 325035, China
| | - Youhong Zhao
- Hepatology Diagnosis and Treatment Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou 325035, China
| | - Zhiyi Chen
- Hepatology Diagnosis and Treatment Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou 325035, China
| | - Lina Wu
- Hepatology Diagnosis and Treatment Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou 325035, China
| | - Sizhe Fang
- Hepatology Diagnosis and Treatment Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou 325035, China
| | - Xiaowei Wang
- Hepatology Diagnosis and Treatment Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou 325035, China
| | - Xiaodong Wang
- Hepatology Diagnosis and Treatment Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou 325035, China
| | - Dazhi Chen
- Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou 325035, China; Hangzhou Medical College, Hangzhou 311300, China.
| | - Yongping Chen
- Hepatology Diagnosis and Treatment Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou 325035, China.
| |
Collapse
|
32
|
Yamashita S, Takase H, Kawakatsu N, Hayashi K, Kin F, Isogaki T, Dohi Y. Fibrosis-4 index is closely associated with future development of hypertension in the Japanese general population. Hypertens Res 2024:10.1038/s41440-024-02028-4. [PMID: 39639128 DOI: 10.1038/s41440-024-02028-4] [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: 06/27/2024] [Revised: 11/07/2024] [Accepted: 11/09/2024] [Indexed: 12/07/2024]
Abstract
The fibrosis-4 index, a noninvasive method for evaluating liver fibrosis, is closely associated with cardiovascular events. In this study, we tested the hypothesis that the fibrosis-4 index is associated with new-onset hypertension in the general population. A total of 15,502 individuals (51.0 ± 13.2 years) who participated in our health checkup program were screened. Participants with hypertension were excluded, and the remaining 8719 normotensive participants (48.4 ± 12.6 years) were followed up (median 1739 days) with the endpoint of the new onset of hypertension. During follow-up, 1750 participants (39.0 per 1000 person-years) developed hypertension. In Kaplan-Meier analysis, where participants were divided into three groups according to the fibrosis-4 index at baseline (low, <1.30; intermediate, 1.30-2.67; high, ≥2.67), the risk of hypertension increased with increasing fibrosis-4 index (low, 33.8; intermediate, 55.2; high, 69.4 per 1000 person-years). Multivariate Cox hazard regression analysis revealed that the log-transformed fibrosis-4 index was independently associated with the development of new hypertension (hazard ratio 4.279, 95% confidence interval 3.318-5.518). These results suggest that the fibrosis-4 index is a useful tool to evaluate a risk of developing hypertension in the general population. Hypertension and liver fibrosis may share a common basis.
Collapse
Affiliation(s)
- Sumiyo Yamashita
- Department of Cardiology, Nagoya City University Mirai Kousei Hospital, Nagoya, Japan
| | - Hiroyuki Takase
- Department of Internal Medicine, Enshu Hospital, Hamamatsu, Japan.
| | - Naomi Kawakatsu
- Department of Internal Medicine, Enshu Hospital, Hamamatsu, Japan
| | - Kazusa Hayashi
- Department of Internal Medicine, Enshu Hospital, Hamamatsu, Japan
| | - Fumihiko Kin
- Department of Internal Medicine, Enshu Hospital, Hamamatsu, Japan
| | - Takeru Isogaki
- Department of Internal Medicine, Enshu Hospital, Hamamatsu, Japan
| | - Yasuaki Dohi
- Division of Internal Medicine, Faculty of Rehabilitation Sciences, Nagoya Gakuin University, Nagoya, Japan
| |
Collapse
|
33
|
Xiang M, Tian X, Wang H, Gan P, Zhang Q. Inappropriate Diet Exacerbates Metabolic Dysfunction-Associated Steatotic Liver Disease via Abdominal Obesity. Nutrients 2024; 16:4208. [PMID: 39683601 DOI: 10.3390/nu16234208] [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: 11/05/2024] [Revised: 11/26/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) represents a refined categorization of non-alcoholic fatty liver disease (NAFLD), highlighting the intricate relationship between hepatic steatosis and metabolic dysfunction. Abdominal obesity (AO), a key diagnostic criterion for metabolic dysfunction, predominantly results from inappropriate diet and unhealthy dietary habits. To comprehensively investigate which dietary factors contribute to MASLD through AO and to understand the underlying biological mechanisms, we initially conducted a systematic review of meta-analysis articles in the PubMed database from the past decade, summarizing dietary factors that affect AO. Subsequently, we conducted targeted searches in the PubMed database for these dietary factors and provided a narrative review of the mechanisms of how these dietary factors lead to AO and how AO exacerbates MASLD. A diet characterized by excessive intake of energy, carbohydrates, fructose, or ultra-processed foods (UPFs) is considered inappropriate. Inappropriate diet leads to the formation of MASLD and AO by enhancing pathways such as de novo lipid synthesis (DNL) in the liver, insulin resistance (IR), gut-liver dysfunction, and inflammation. Dietary interventions for inappropriate diets can effectively intervene in and improve MASLD and AO. The mechanism of inappropriate diet on abdominal fat deposition is through excessive energy or the activation of the enzyme 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD-1) to increase endocortisol secretion. Then, the excessive accumulation of visceral fat facilitates a rapid and augmented flux of free fatty acids (FFAs) to the liver and initiates a series of deleterious effects, including oxidative stress (OS), endoplasmic reticulum stress (ERS), activation of protein kinase C (PKC) pathways, and inflammation. Additionally, FFAs may mediate excessive lipid deposition and hepatocellular damage through the action of hormones. These pathways to liver damage exacerbate MASLD and progression to metabolic dysfunction-associated steatohepatitis (MASH) and fibrosis. Furthermore, investigating other potential mechanisms by which AO may influence MASLD could offer new recommendations for the treatment guidelines of MASLD.
Collapse
Affiliation(s)
- Minghui Xiang
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Xiaoli Tian
- School of Public Health, Xinjiang Medical College, Ürümqi 830000, China
- School of Public Health, Xinjiang Second Medical College, Karamay 834000, China
| | - Hui Wang
- Department of Maternal and Child Health, School of Public Health, Peking University, Beijing 100191, China
| | - Ping Gan
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou 511400, China
| | - Qian Zhang
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| |
Collapse
|
34
|
Lee EO, Jin H, Kim S, Joo HK, Lee YR, An SY, Piao S, Lee KH, Jeon BH. Alterations in Adipose Tissue and Adipokines in Heterozygous APE1/Ref-1 Deficient Mice. Endocrinol Metab (Seoul) 2024; 39:932-945. [PMID: 39566547 PMCID: PMC11695485 DOI: 10.3803/enm.2024.2061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/29/2024] [Accepted: 08/19/2024] [Indexed: 11/22/2024] Open
Abstract
BACKGRUOUND The role of apurinic/apyrimidinic endonuclease 1/redox factor-1 (APE1/Ref-1) in adipose tissue remains poorly understood. This study investigates adipose tissue dysfunction in heterozygous APE1/Ref-1 deficiency (APE1/Ref-1+/-) mice, focusing on changes in adipocyte physiology, oxidative stress, adipokine regulation, and adipose tissue distribution. METHODS APE1/Ref-1 mRNA and protein levels in white adipose tissue (WAT) were measured in APE1/Ref-1+/- mice, compared to their wild-type (APE1/Ref-1+/+) controls. Oxidative stress was assessed by evaluating reactive oxygen species (ROS) levels. Histological and immunohistochemical analyses were conducted to observe adipocyte size and macrophage infiltration of WAT. Adipokine expression was measured, and micro-magnetic resonance imaging (MRI) was used to quantify abdominal fat volumes. RESULTS APE1/Ref-1+/- mice exhibited significant reductions in APE1/Ref-1 mRNA and protein levels in WAT and liver tissue. These mice also showed elevated ROS levels, suggesting a regulatory role for APE1/Ref-1 in oxidative stress in WAT and liver. Histological and immunohistochemical analyses revealed hypertrophic adipocytes and macrophage infiltration in WAT, while Oil Red O staining demonstrated enhanced ectopic fat deposition in the liver of APE1/Ref-1+/- mice. These mice also displayed altered adipokine expression, with decreased adiponectin and increased leptin levels in the WAT, along with corresponding alterations in plasma levels. Despite no significant changes in overall body weight, microMRI assessments demonstrated a significant increase in visceral and subcutaneous abdominal fat volumes in APE1/Ref-1+/- mice. CONCLUSION APE1/Ref-1 is crucial in adipokine regulation and mitigating oxidative stress. These findings suggest its involvement in adipose tissue dysfunction, highlighting its potential impact on abdominal fat distribution and its implications for obesity and oxidative stress-related conditions.
Collapse
Affiliation(s)
- Eun-Ok Lee
- Research Institute of Medical Sciences, Chungnam National University College of Medicine, Daejeon, Korea
- Department of Physiology, Chungnam National University College of Medicine, Daejeon, Korea
| | - Hao Jin
- Department of Physiology, Chungnam National University College of Medicine, Daejeon, Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Korea
| | - Sungmin Kim
- Department of Physiology, Chungnam National University College of Medicine, Daejeon, Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Korea
| | - Hee Kyoung Joo
- Research Institute of Medical Sciences, Chungnam National University College of Medicine, Daejeon, Korea
- Department of Physiology, Chungnam National University College of Medicine, Daejeon, Korea
| | - Yu Ran Lee
- Research Institute of Medical Sciences, Chungnam National University College of Medicine, Daejeon, Korea
- Department of Physiology, Chungnam National University College of Medicine, Daejeon, Korea
| | - Soo Yeon An
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Korea
- Division of Cardiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Shuyu Piao
- Research Institute of Medical Sciences, Chungnam National University College of Medicine, Daejeon, Korea
- Department of Physiology, Chungnam National University College of Medicine, Daejeon, Korea
| | - Kwon Ho Lee
- Department of Physical Therapy, Joongbu University, Geumsan, Korea
| | - Byeong Hwa Jeon
- Research Institute of Medical Sciences, Chungnam National University College of Medicine, Daejeon, Korea
- Department of Physiology, Chungnam National University College of Medicine, Daejeon, Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Korea
| |
Collapse
|
35
|
Yadav P, Quadri K, Kadian R, Waziri A, Agrawal P, Alam MS. New approaches to the treatment of metabolic dysfunction-associated steatotic liver with natural products. ILIVER 2024; 3:100131. [DOI: 10.1016/j.iliver.2024.100131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
36
|
Sattari M, Amri J, Shahaboddin ME, Sattari M, Tabatabaei-Malazy O, Azmon M, Meshkani R, Panahi G. The protective effects of fisetin in metabolic disorders: a focus on oxidative stress and associated events. J Diabetes Metab Disord 2024; 23:1753-1771. [PMID: 39610486 PMCID: PMC11599505 DOI: 10.1007/s40200-024-01502-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 09/09/2024] [Indexed: 11/30/2024]
Abstract
Abstract Metabolic syndrome is increasingly recognized as a significant precursor to various chronic diseases, contributing to a growing public health concern. Its complex pathogenesis involves multiple interrelated mechanisms, with oxidative stress identified as a cornerstone that exacerbates other pathogenic pathways. This study elucidates the molecular mechanisms by which oxidative stress intensifies metabolic disturbances, particularly insulin resistance. Some recent research has focused on fisetin, a natural product known for its potential benefits in diabetes and its associated microvascular and macrovascular complications. This paper compiles a comprehensive collection of findings by reviewing studies conducted over the past decade, detailing dosages, investigated markers, and their respective outcomes. Notably, a recurrent finding was fisetin's ability to enhance Nrf2, a principal regulator of antioxidant defense, in both metabolic and non-metabolic diseases. Furthermore, intriguing results suggest that the effects of Nrf2 extend beyond oxidative stress modulation, demonstrating favorable impacts on tissue-specific functions in metabolic regulation. This highlights fisetin not only as an antioxidant but also as a potential therapeutic agent for improving metabolic health and mitigating the effects of metabolic syndrome. In conclusion, fisetin can enhance the body's antioxidant defenses by modulating the Nrf2 pathway while also improving metabolic health through its effects on inflammation, cell survival, and energy metabolism, offering a comprehensive approach to managing metabolic disorders. Graphical Abstract
Collapse
Affiliation(s)
- Mahboobe Sattari
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Students’ Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, I.R Iran
| | - Jamal Amri
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Students’ Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, I.R Iran
| | - Mohammad Esmaeil Shahaboddin
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohadese Sattari
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Ozra Tabatabaei-Malazy
- Non-Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Marzyeh Azmon
- Department of Internal Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reza Meshkani
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ghodratollah Panahi
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
37
|
Mrwad AA, El-Shafey SE, Said NM. Chitosan-encapsulated selenium nanoparticles alleviate CCl 4 induced hepatotoxicity through synergistically modulating NF-κB and Nrf2 signaling pathways and regulating Bcl-2 and Caspase-3 expression: A comprehensive study with multiple regression analysis. J Trace Elem Med Biol 2024; 86:127563. [PMID: 39547053 DOI: 10.1016/j.jtemb.2024.127563] [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: 08/17/2024] [Revised: 10/24/2024] [Accepted: 11/08/2024] [Indexed: 11/17/2024]
Abstract
BACKGROUND The delivery of selenium in a nano-form (Se-NPs) is a promising modality of treatment for various oxidative stress-induced diseases. OBJECTIVE This study aims to investigate the conceivable effects of selenium nanoparticles either alone (Se-NPs) or encapsulated with chitosan (Se-CS-NPs) on toxicity induced by CCl4 in rats. METHODS Eighty albino rats were divided equally into eight groups. The first group was the placebo. The second group was a positive control, while the third and the fourth groups got orally (Se-NPs 5 mg/Kg) and (Se-CS-NPs 225 mg/Kg) respectively. The fifth and sixth groups were protective groups in which Se-NPs or Se-CS-NPs were given simultaneously. The seventh and eighth groups were therapeutic as they received either Se-NPs or Se-CS-NPs after stopping the CCl4 injection for 4 weeks more. RESULTS Our results showed that the protective and therapeutic groups showed an increase in caspase-3 gene expression with a decline in the expression of Bcl-2, Nrf2, and AFP genes. Histopathological and immunohistochemical investigations showed the role of selenium nanoparticles either alone or coated with chitosan in decreasing fibrotic marker collagen I positive reaction CONCLUSION: Selenium nanoparticles showed an excellent effect in counteracting the toxic effect of carbon tetrachloride on liver functions, inflammation reactions, and apoptosis process. Moreover, using selenium nanoparticles has a strong role in preserving the liver architecture with its normal constituents. No additional benefit was observed when the selenium nanoparticles were encapsulated with chitosan.
Collapse
Affiliation(s)
| | - Shaymaa E El-Shafey
- Physical Chemistry Department, Surface and Catalysis Lab., National Research Center, El-Bohouth St. 33, Dokki, Giza, Egypt
| | - Noha Mohamed Said
- Biochemistry Department, Faculty of Science, Zagazig University, Zagazig 44519, Egypt.
| |
Collapse
|
38
|
Lin Y, Ye H, Chen Y, Zhang R, Chen Y, Ou W. Integrative Analyses of Genes of Pediatric Non-alcoholic Fatty Liver Disease Associated with Energy Metabolism. Dig Dis Sci 2024; 69:4373-4391. [PMID: 39496907 PMCID: PMC11602812 DOI: 10.1007/s10620-024-08702-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 10/18/2024] [Indexed: 11/06/2024]
Abstract
BACKGROUND Pediatric non-alcoholic fatty liver disease (NAFLD) is a chronic steatosis of the liver associated with energy metabolism in children and adolescents, failure to intervene promptly can elevate the risk of developing hepatocellular carcinoma. Therefore, this study aimed to understand the underlying mechanism of pediatric NAFLD and investigate potential biomarkers and therapeutic targets. METHODS We investigated genes using the GSE185051 data set related to energy metabolism from the GeneCards database, constructed protein-protein interaction network, identified hub genes and established networks representing interactions between these hub genes and miRNA, RNA-binding proteins, transcription factors, and drugs. Subsequently, we performed Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) functional enrichment analysis, Gene Set Enrichment Analysis (GSEA), and immune infiltration analysis. RESULTS Our analysis identified 9 hub genes through the PPI network. The target molecules were identified through the interaction network between hub genes and miRNAs, RNA-binding proteins, transcription factors, and drugs. GO analysis revealed that hub genes were associated with oxidative stress responses and other pathways. KEGG analysis highlighted their involvement in pathways such as insulin resistance, among others. GSEA revealed that hub genes were highly enriched in pathways related to Omega-9 fatty acid synthesis, among others. Immune infiltration analysis suggested that mast cells and T follicular helper cells play significant roles in the pathogenesis of NAFLD. CONCLUSION We identified the hub genes in pediatric NAFLD closely related to energy metabolism. These findings offer the potential for identifying potential novel diagnostic biomarkers, and establishing therapeutic targets for pediatric NAFLD.
Collapse
Affiliation(s)
- Yijun Lin
- Department of Pediatrics, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, No.18, Daoshan Road, Gulou District, Fuzhou, 350001, Fujian, China
- Department of Pediatrics, Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), Fuzhou, China
| | - Hong Ye
- Department of Pediatrics, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, No.18, Daoshan Road, Gulou District, Fuzhou, 350001, Fujian, China.
- Department of Pediatrics, Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), Fuzhou, China.
| | - Yan Chen
- Department of Pediatrics, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, No.18, Daoshan Road, Gulou District, Fuzhou, 350001, Fujian, China
- Department of Pediatrics, Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), Fuzhou, China
| | - Rui Zhang
- Department of Pediatrics, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, No.18, Daoshan Road, Gulou District, Fuzhou, 350001, Fujian, China
- Department of Pediatrics, Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), Fuzhou, China
| | - Yuyun Chen
- Department of Pediatrics, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, No.18, Daoshan Road, Gulou District, Fuzhou, 350001, Fujian, China
- Department of Pediatrics, Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), Fuzhou, China
| | - Weijie Ou
- Department of Pediatrics, Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), Fuzhou, China
| |
Collapse
|
39
|
Chen J, Lu RS, Diaz-Canestro C, Song E, Jia X, Liu Y, Wang C, Cheung CK, Panagiotou G, Xu A. Distinct changes in serum metabolites and lipid species in the onset and progression of NAFLD in Obese Chinese. Comput Struct Biotechnol J 2024; 23:791-800. [PMID: 38318437 PMCID: PMC10839226 DOI: 10.1016/j.csbj.2024.01.007] [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: 08/28/2023] [Revised: 01/08/2024] [Accepted: 01/14/2024] [Indexed: 02/07/2024] Open
Abstract
Introduction Metabolic disturbances are major contributors to the onset and progression of non-alcoholic fatty liver disease (NAFLD), which includes a histological spectrum ranging from single steatosis (SS) to non-alcoholic steatohepatitis (NASH). This study aimed to identify serum metabolites and lipids enriched in different histological stages of NAFLD and to explore metabolites/lipids as non-invasive biomarkers in risk prediction of NAFLD and NASH in obese Chinese. Methods Serum samples and liver biopsies were obtained from 250 NAFLD subjects. Untargeted metabolomic and lipidomic profiling were performed using Liquid Chromatography-Mass Spectrometry. Significantly altered metabolites and lipids were identified by MaAsLin2. Pathway enrichment was conducted with MetaboAnalyst and LIPEA. WGCNA was implemented to construct the co-expression network. Logistic regression models were developed to classify different histological stages of NAFLD. Results A total of 263 metabolites and 550 lipid species were detected in serum samples. Differential analysis and pathway enrichment analysis revealed the progressive patterns in metabolic mechanisms during the transition from normal liver to SS and to NASH, including N-palmitoyltaurine, tridecylic acid, and branched-chain amino acid signaling pathways. The co-expression network showed a distinct correlation between different triglyceride and phosphatidylcholine species with disease severity. Multiple models classifying NAFLD versus normal liver and NASH versus SS identified important metabolic features associated with significant improvement in disease prediction compared to conventional clinical parameters. Conclusion Different histological stages of NAFLD are enriched with distinct sets of metabolites, lipids, and metabolic pathways. Integrated algorithms highlight the important metabolic and lipidomic features for diagnosis and staging of NAFLD in obese individuals.
Collapse
Affiliation(s)
- Jiarui Chen
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong Special Administrative Region
- Department of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region
- Leibniz Insitute for Natural Product Research and Infection Biology, Microbiome Dynamics, Hans Knöll Institute, Jena, Germany
| | - Ronald Siyi Lu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong Special Administrative Region
- Department of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Candela Diaz-Canestro
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong Special Administrative Region
- Department of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Erfei Song
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong Special Administrative Region
- Department of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region
- Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xi Jia
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong Special Administrative Region
- Department of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Yan Liu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong Special Administrative Region
- Department of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Cunchuan Wang
- Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Cynthia K.Y. Cheung
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong Special Administrative Region
- Department of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Gianni Panagiotou
- Department of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region
- Leibniz Insitute for Natural Product Research and Infection Biology, Microbiome Dynamics, Hans Knöll Institute, Jena, Germany
- Friedrich Schiller University, Faculty of Biological Sciences, Jena, Germany
- Cluster of Excellence Balance of the Microverse, Friedrich-Schiller-University Jena, Jena, Germany
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong Special Administrative Region
- Department of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region
- Department of Pharmacology and Pharmacy, the University of Hong Kong, Hong Kong Special Administrative Region
| |
Collapse
|
40
|
Li J, Lv M, Yuan Z, Ge J, Geng T, Gong D, Zhao M. PGC-1α Promotes mitochondrial biosynthesis and energy metabolism of goose fatty liver. Poult Sci 2024; 104:104617. [PMID: 39644719 DOI: 10.1016/j.psj.2024.104617] [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: 09/19/2024] [Revised: 11/15/2024] [Accepted: 11/30/2024] [Indexed: 12/09/2024] Open
Abstract
To investigate the functions of peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) in the goose fatty liver, a total of 30 healthy 63-day-old male Landes geese were selected and randomly assigned to control group and overfeeding group. The overexpression or RNA interference assay of PGC-1α was performed in goose primary hepatocytes. Our data showed that the PGC-1α expression was increased in fatty liver. The abundance of mitochondrial biosynthesis-related and energy metabolism-related genes, including mitochondrial transcription factor A (TFAM), mitochondrial transcription factor B1 (TFB1M), mitochondrial transcription factor B2 (TFB2M), nuclear respiratory factor 1 (NRF1), DNA topoisomerase I mitochondrial (TOP1MT), peroxisome proliferator-activated receptor gamma coactivator 1-beta (PGC-1β), sirtuin 3 (SIRT3), mitochondrially encoded cytochrome B (CYTB), and AMP-activated protein kinase alpha (AMPKα) were significantly increased in fatty liver. The abundance of TFAM, TFB1M, TFB2M, NRF1, and TOP1MT transcript was induced by PGC-1α overexpression, but inhibited by PGC-1α interference in primary hepatocytes. The mRNA expression levels of PGC-1β, SIRT3, SIRT5, CYTB, and AMPKα were significantly enhanced after PGC-1α overexpression. However, the mRNA expression levels of PGC-1β, SIRT5 and AMPKα were decreased after PGC-1α interference. Furthermore, we observed a significant increase in the mitochondrial DNA (mtDNA) copy number, the activity of mitochondrial respiratory chain complex Ⅳ (MRCC Ⅳ), succinate dehydrogenase (SDH), malate dehydrogenase (MDH), and the NAD+/NADH ratio in fatty liver. But the activity of MRCC Ⅴ, as well as the levels of ADP and ATP in fatty liver were reduced. Additionally, the mtDNA copy number, the activity of MRCC Ⅰ, MRCC Ⅲ-Ⅴ, SDH, and MDH, and NAD+/NADH ratio were enhanced by PGC-1α overexpression; Whereas the mtDNA copy number, the activity of MRCC Ⅰ, SDH, and MDH, and the ratio of NAD+/NADH were inhibited by PGC-1α interference. In conclusion, these findings suggest that PGC-1α improves mitochondrial biosynthesis and energy metabolism in goose fatty liver, which may be an adaptive mechanism for goose fatty liver to cope with steatosis.
Collapse
Affiliation(s)
- Jiahui Li
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu Province 225009, PR China
| | - Mengqing Lv
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu Province 225009, PR China
| | - Zijin Yuan
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu Province 225009, PR China
| | - Jing Ge
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu Province 225009, PR China
| | - Tuoyu Geng
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu Province 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu Province 225009, PR China
| | - Daoqing Gong
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu Province 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu Province 225009, PR China
| | - Minmeng Zhao
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu Province 225009, PR China.
| |
Collapse
|
41
|
Alvarado-Tapias E, Maya-Miles D, Albillos A, Aller R, Ampuero J, Andrade RJ, Arechederra M, Aspichueta P, Banales JM, Blas-García A, Caparros E, Cardoso Delgado T, Carrillo-Vico A, Claria J, Cubero FJ, Díaz-Ruiz A, Fernández-Barrena MG, Fernández-Iglesias A, Fernández-Veledo S, Francés R, Gallego-Durán R, Gracia-Sancho J, Irimia M, Lens S, Martínez-Chantar ML, Mínguez B, Muñoz-Hernández R, Nogueiras R, Ramos-Molina B, Riveiro-Barciela M, Rodríguez-Perálvarez ML, Romero-Gómez M, Sabio G, Sancho-Bru P, Ventura-Cots M, Vidal S, Gahete MD. Proceedings of the 5th Meeting of Translational Hepatology, organized by the Spanish Association for the Study of the Liver (AEEH). GASTROENTEROLOGIA Y HEPATOLOGIA 2024; 47:502207. [PMID: 38723772 DOI: 10.1016/j.gastrohep.2024.502207] [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: 04/15/2024] [Accepted: 05/02/2024] [Indexed: 11/30/2024]
Abstract
This is the summary report of the 5th Translational Hepatology Meeting, endorsed by the Spanish Association for the Study of the Liver (AEEH) and held in Seville, Spain, in October 2023. The meeting aimed to provide an update on the latest advances in the field of basic and translational hepatology, covering different molecular, cellular, and pathophysiological aspects of the most relevant clinical challenges in liver pathologies. This includes the identification of novel biomarkers and diagnostic tools, the understanding of the relevance of immune response and inflammation in liver diseases, the characterization of current medical approaches to reverse liver diseases, the incorporation of novel molecular insights through omics techniques, or the characterization of the impact of toxic and metabolic insults, as well as other organ crosstalk, in liver pathophysiology.
Collapse
Affiliation(s)
- Edilmar Alvarado-Tapias
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Department of Gastroenterology, Hospital Santa Creu I Sant Pau, Institut de Recerca Sant Pau, Universidad Autónoma de Barcelona, Barcelona, Spain.
| | - Douglas Maya-Miles
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío (HUVR), CISC, Universidad de Sevilla, Sevilla, Spain.
| | - Agustin Albillos
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Servicio de Gastroenterología y Hepatología, Hospital Universitario Ramón y Cajal/Universidad de Alcalá/Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Rocio Aller
- BioCritic, Group for Biomedical Research in Critical Care Medicine, Spain; Department of Medicine, Dermatology and Toxicology, Universidad de Valladolid, Spain; Gastroenterology Unit, Hospital Clínico Universitario de Valladolid, 47003 Valladolid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain; Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga, Spain
| | - Javier Ampuero
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío (HUVR), CISC, Universidad de Sevilla, Sevilla, Spain
| | - Raul J Andrade
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga, Spain
| | - Maria Arechederra
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Hepatology Laboratory, Solid Tumors Program, CIMA, CCUN, University of Navarra, Pamplona, Spain; Instituto de Investigaciones Sanitarias de Navarra IdiSNA, Pamplona, Spain
| | - Patricia Aspichueta
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, Leioa, Spain; Biobizkaia Health Research Institute, Barakaldo, Spain
| | - Jesus M Banales
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Department of Liver and Gastrointestinal Diseases, Biogipuzkoa Health Research Institute - Donostia University Hospital - University of the Basque Country (UPV/EHU), Ikerbasque, Donostia-San Sebastian, Spain; Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
| | - Ana Blas-García
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Departamento de Fisiología, Universitat de València, Av. Blasco Ibáñez, 15, 46010 Valencia, Spain; FISABIO (Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana), Av. de Catalunya, 21, 46020 Valencia, Spain
| | - Esther Caparros
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Grupo de Inmunobiología Hepática e Intestinal, Departamento Medicina Clínica, Universidad Miguel Hernández, San Juan, Spain; Instituto de Investigación Sanitaria ISABIAL, Hospital General Universitario de Alicante, Alicante, Spain
| | - Teresa Cardoso Delgado
- Biobizkaia Health Research Institute, Barakaldo, Spain; IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Antonio Carrillo-Vico
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío (HUVR), CISC, Universidad de Sevilla, Sevilla, Spain; Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Facultad de Medicina, Universidad de Sevilla, Seville, Spain
| | - Joan Claria
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Biochemistry and Molecular Genetics Service, Hospital Clínic, IDIBAPS, Barcelona, Spain; University of Barcelona, Spain
| | - Francisco Javier Cubero
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, Madrid, Spain; Health Research Institute Gregorio Marañón (IiSGM), Madrid, Spain
| | - Alberto Díaz-Ruiz
- Laboratory of Cellular and Molecular Gerontology, Precision Nutrition and Aging, Madrid Institute for Advanced Studies - IMDEA Food, CEI UAM+CSIC, Madrid, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Madrid, Spain
| | - Maite G Fernández-Barrena
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Instituto de Investigaciones Sanitarias de Navarra IdiSNA, Pamplona, Spain; Hepatology Laboratory, Solid Tumors Program, CIMA, CCUN, University of Navarra, Spain
| | - Anabel Fernández-Iglesias
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Liver Vascular Biology Research Group, IDIBAPS, Hospital Clínic de Barcelona, 08036 Barcelona, Spain
| | - Sonia Fernández-Veledo
- Department of Endocrinology and Nutrition and Research Unit, University Hospital of Tarragona Joan XXIII, Institut d'Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili (URV), Tarragona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Ruben Francés
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Grupo de Inmunobiología Hepática e Intestinal, Departamento Medicina Clínica, Universidad Miguel Hernández, San Juan, Spain; Instituto de Investigación Sanitaria ISABIAL, Hospital General Universitario de Alicante, Alicante, Spain
| | - Rocío Gallego-Durán
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío (HUVR), CISC, Universidad de Sevilla, Sevilla, Spain
| | - Jordi Gracia-Sancho
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Liver Vascular Biology Research Group, IDIBAPS, Hospital Clínic de Barcelona, 08036 Barcelona, Spain
| | - Manuel Irimia
- Universitat Pompeu Fabra (UPF), Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, ICREA, Barcelona, Spain
| | - Sabela Lens
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, Madrid, Spain; Liver Unit, Hospital Clínic, IDIBAPS, Barcelona, Spain
| | - María Luz Martínez-Chantar
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Liver Disease Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Bizkaia, Spain
| | - Beatriz Mínguez
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Liver Unit, Hospital Universitari Vall d'Hebron, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Rocío Muñoz-Hernández
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío (HUVR), CISC, Universidad de Sevilla, Sevilla, Spain; Departamento de fisiología, Facultad de Biología, Universidad de Sevilla, Sevilla, Spain
| | - Rubén Nogueiras
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Madrid, Spain; Department of Physiology, CIMUS, University of Santiago de Compostela, Instituto de Investigación Sanitaria, Santiago de Compostela, Spain; Galician Agency of Innovation (GAIN), Xunta de Galicia, Santiago de Compostela, Spain
| | - Bruno Ramos-Molina
- Obesity, Diabetes and Metabolism Laboratory, Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| | - Mar Riveiro-Barciela
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Liver Unit, Hospital Universitari Vall d'Hebron, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Manuel L Rodríguez-Perálvarez
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Department of Hepatology and Liver Transplantation, Reina Sofia University Hospital, Cordoba, Spain; Maimonides Biomedical Research Institute of Córdoba (IMIBIC), University of Córdoba, Cordoba, Spain
| | - Manuel Romero-Gómez
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío (HUVR), CISC, Universidad de Sevilla, Sevilla, Spain
| | - Guadalupe Sabio
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Stress Kinases in Diabetes, Cancer and Biochemistry, Spain; Centro Nacional de Investigaciones Oncologicas (CNIO), Organ Crosstalk in Metabolic Diseases, Madrid, Spain
| | - Pau Sancho-Bru
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Meritxell Ventura-Cots
- Liver Unit, Hospital Universitari Vall d'Hebron, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain; Center for Liver Diseases, Pittsburgh Liver Research Center, Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Silvia Vidal
- Group of Inflammatory Diseases, Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
| | - Manuel D Gahete
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Madrid, Spain; Department of Cell Biology, Physiology and Immunology, University of Córdoba, Spain; Molecular Hepatology Group, Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Spain; Reina Sofia University Hospital, Cordoba, Spain.
| |
Collapse
|
42
|
Zhang X, Yin G, Chen S, Meng D, Yu W, Liu H, Wang L, Zhang F. Diosgenin ameliorating non-alcoholic fatty liver disease via Nrf2-mediated regulation of oxidative stress and ferroptosis. Diabetes Obes Metab 2024; 26:5745-5756. [PMID: 39344834 DOI: 10.1111/dom.15945] [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: 06/06/2024] [Revised: 08/17/2024] [Accepted: 08/29/2024] [Indexed: 10/01/2024]
Abstract
AIM This study aimed to investigate the mechanisms through which diosgenin inhibits the pathogenesis of non-alcoholic fatty liver disease, focusing particularly on ferroptosis-related pathways and its reliance on nuclear factor erythroid 2-related factor 2. MATERIALS AND METHODS Using a rat model, we showed diosgenin's efficacy in reducing lipid deposition throughout the body and examined its impact on ferroptosis-related gene expression in vivo. Moreover, in vitro experiments using human hepatocellular liver carcinoma cell line cells were conducted to assess oxidative stress and ferroptosis levels. RESULTS Diosgenin decreased lipid accumulation and steatosis; lowered serum levels of total cholesterol, triglycerides, low-density lipoprotein cholesterol, glutamic pyruvic transaminase and glutamic oxaloacetic transaminase; reduced interleukin-1β and tumour necrosis factor-α; diosgenin decreased malondialdehyde levels; and increased serum superoxide dismutase levels in a rat model of high-fat diet-induced non-alcoholic fatty liver disease. Diosgenin upregulated the expression of nuclear factor erythroid 2-related factor 2 and its downstream ferroptosis-related genes to inhibit ferroptosis in the livers of rats with non-alcoholic fatty liver disease. Diosgenin decreased reactive oxygen species levels and enhanced the expression of ferroptosis-related genes in human hepatocellular liver carcinoma cells induced by free fatty acids, with its effects being dependent on nuclear factor erythroid 2-related factor 2. CONCLUSIONS This study highlights the potential of diosgenin from Dioscoreaceae plants in mitigating oxidative stress and ferroptosis levels through nuclear factor erythroid 2-related factor 2 regulation, offering novel insights into the treatment of non-alcoholic fatty liver disease and other metabolic disorders through traditional Chinese medicine.
Collapse
Affiliation(s)
- Xin Zhang
- Department of The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Guoliang Yin
- Department of The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Suwen Chen
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Decheng Meng
- Department of The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wenfei Yu
- Department of The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Hongshuai Liu
- Department of The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Linya Wang
- Department of The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Fengxia Zhang
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
43
|
Chen C, Zhou X, Cheng W, Li X, Zhang B, Tu J, Meng J, Peng Y, Duan X, Yu Q, Tan X. Design, synthesis and FXR partial agonistic activity of anthranilic acid derivatives bearing aryloxy moiety as therapeutic agents for metabolic dysfunction-associated steatohepatitis. Bioorg Chem 2024; 153:107940. [PMID: 39515132 DOI: 10.1016/j.bioorg.2024.107940] [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: 09/03/2024] [Revised: 10/26/2024] [Accepted: 11/02/2024] [Indexed: 11/16/2024]
Abstract
Farnesoid X receptor (FXR) is considered a promising therapeutic target for the treatment of metabolic dysfunction-associated steatohepatitis (MASH). Increasing evidence suggests that targeting FXR with full agonists may lead to side effects. FXR partial agonists, which moderately activate FXR signaling, are emerging as a feasible approach to mitigate side effects and address MASH. Herein, a series of novel anthranilic acid derivatives bearing aryloxy moiety were designed and synthesized using a hybrid strategy from the previously identified FXR partial agonists DM175 and AIV-25. Particularly, compound 26 exhibited potent FXR partial agonistic activity in a dual-luciferase reporter gene assay with an EC50 value of 0.09 ± 0.02 µM (75.13 % maximum efficacy relative to OCA). In the MASH mice model, compound 26 significantly ameliorated the pathological features of the liver, including steatosis, inflammation, and fibrosis. In addition, compound 26 displayed high selectivity, good oral bioavailability, high liver distribution, as well as an acceptable safety profile. Molecular simulation studies showed that compound 26 fitted well with the binding site of FXR. Collectively, these findings demonstrated that compound 26 might serve as a promising candidate targeting FXR for MASH treatment.
Collapse
Affiliation(s)
- Cong Chen
- Guangxi Key Laboratory of Drug Discovery and Optimization, College of Pharmacy, Guilin Medical University, Guilin 541199, China; Guangxi Key Laboratory of Environmental Exposure Omics and Life Cycle Health, College of Public Health, Guilin Medical University, Guilin 541199, China
| | - Xianghui Zhou
- Guangxi Key Laboratory of Drug Discovery and Optimization, College of Pharmacy, Guilin Medical University, Guilin 541199, China; Department of Pharmacy, Yunfu People's Hospital, Yunfu 527300, China
| | - Wa Cheng
- Guangxi Key Laboratory of Drug Discovery and Optimization, College of Pharmacy, Guilin Medical University, Guilin 541199, China
| | - Xin Li
- School of Life Sciences, Guangzhou University, Guangzhou 510006, China
| | - Bing Zhang
- Guangxi Key Laboratory of Drug Discovery and Optimization, College of Pharmacy, Guilin Medical University, Guilin 541199, China
| | - Jiaojiao Tu
- Guangxi Key Laboratory of Drug Discovery and Optimization, College of Pharmacy, Guilin Medical University, Guilin 541199, China
| | - Jieyun Meng
- Guangxi Key Laboratory of Drug Discovery and Optimization, College of Pharmacy, Guilin Medical University, Guilin 541199, China
| | - Yanfen Peng
- Guangxi Key Laboratory of Drug Discovery and Optimization, College of Pharmacy, Guilin Medical University, Guilin 541199, China
| | - Xiaoqun Duan
- Guangxi Key Laboratory of Drug Discovery and Optimization, College of Pharmacy, Guilin Medical University, Guilin 541199, China.
| | - Qiming Yu
- Guangxi Key Laboratory of Environmental Exposure Omics and Life Cycle Health, College of Public Health, Guilin Medical University, Guilin 541199, China.
| | - Xiangduan Tan
- Guangxi Key Laboratory of Drug Discovery and Optimization, College of Pharmacy, Guilin Medical University, Guilin 541199, China.
| |
Collapse
|
44
|
Jang CH, Chung YC, Lee A, Hwang YH. Hydroethanolic Extract of Polygonum aviculare L. Mediates the Anti-Inflammatory Activity in RAW 264.7 Murine Macrophages Through Induction of Heme Oxygenase-1 and Inhibition of Inducible Nitric Oxide Synthase. PLANTS (BASEL, SWITZERLAND) 2024; 13:3314. [PMID: 39683107 DOI: 10.3390/plants13233314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 11/07/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024]
Abstract
Polygonum aviculare L. (PAL), commonly known as knotgrass, has been utilized as a traditional folk medicine across Asian, African, Latin American and Middle Eastern countries to treat various inflammatory diseases, including arthritis and airway inflammation. Numerous medicinal herbs exert anti-inflammatory and antioxidative effects that are mediated through the activation of nuclear factor-erythroid 2-related factor 2 (Nrf2) and the inhibition of nuclear factor kappa B (NF-κB). However, the underlying molecular mechanisms linking the antioxidative and anti-inflammatory effects remain poorly understood. Heme oxygenase-1 (HO-1) is an antioxidant enzyme that catalyzes heme degradation, ultimately leading to the production of carbon monoxide (CO). Elevated levels of CO have been correlated with the decreased level of inducible nitric oxide synthase (iNOS). In this study, we examined whether HO-1 plays a key role in the relationship between the antioxidative and anti-inflammatory properties of PAL. The anti-inflammatory and antioxidative activities of PAL in an in vitro system were evaluated by determining NF-κB activity, antioxidant response element (ARE) activity, pro-inflammatory cytokine and protein levels, as well as antioxidant protein levels. To examine whether HO-1 inhibition interfered with the anti-inflammatory effect of PAL, we measured nitrite, reactive oxygen species, iNOS, and HO-1 levels in RAW 264.7 murine macrophages pre-treated with Tin protoporphyrin (SnPP, an HO-1 inhibitor). Our results demonstrated that PAL increased ARE activity and the Nrf2-regulated HO-1 level, exerting antioxidative activities in RAW 264.7 macrophages. Additionally, PAL reduced cyclooxygenase-2 (COX-2) and iNOS protein levels by inactivating NF-κB in lipopolysaccharide (LPS)-activated RAW 264.7 macrophages. Further investigation using the HO-1 inhibitor revealed that HO-1 inhibition promoted iNOS expression, subsequently elevating nitric oxide (NO) generation in LPS-activated RAW 264.7 macrophages treated with PAL compared to those in the macrophages without the HO-1 inhibitor. Overall, our findings suggest that HO-1 induction by PAL may exert anti-inflammatory effects through the reduction of the iNOS protein level. Hence, this study paves the way for further investigation to understand molecular mechanisms underlying the antioxidative and anti-inflammatory activities of medicinal herbs.
Collapse
Affiliation(s)
- Chan Ho Jang
- Herbal Medicine Research Division, Korea Institution of Oriental Medicine (KIOM), Daejeon 34054, Republic of Korea
| | - You Chul Chung
- Herbal Medicine Research Division, Korea Institution of Oriental Medicine (KIOM), Daejeon 34054, Republic of Korea
| | - Ami Lee
- Herbal Medicine Research Division, Korea Institution of Oriental Medicine (KIOM), Daejeon 34054, Republic of Korea
- Korean Convergence Medical Science Major, KIOM School, University of Science & Technology (UST), Daejeon 34054, Republic of Korea
| | - Youn-Hwan Hwang
- Herbal Medicine Research Division, Korea Institution of Oriental Medicine (KIOM), Daejeon 34054, Republic of Korea
- Korean Convergence Medical Science Major, KIOM School, University of Science & Technology (UST), Daejeon 34054, Republic of Korea
| |
Collapse
|
45
|
Xu Y, Yan Z, Liu L. Association between advanced lung cancer inflammation index and osteoporosis in patients with type 2 diabetes mellitus: evidence from NHANES. Front Endocrinol (Lausanne) 2024; 15:1421696. [PMID: 39655346 PMCID: PMC11625538 DOI: 10.3389/fendo.2024.1421696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 11/04/2024] [Indexed: 12/12/2024] Open
Abstract
Background Previous studies have shown a significantly increased prevalence of osteoporosis (OP) in patients with type 2 diabetes mellitus (T2DM), which is closely associated with inflammation and nutrition. This study aimed to investigate the relationship between the advanced lung cancer inflammation index (ALI) and OP in patients with T2DM. Methods This cross-sectional analysis was conducted based on data from middle-aged and older adults aged 50 years and older with T2DM from the National Health and Nutrition Examination Survey (NHANES).Weighted multivariable logistic regression and linear regression were utilized to investigate the correlation between the ALI and OP with femur bone mineral density (BMD) in individuals with T2DM. Restricted cubic splines (RCS) were employed to assess potential nonlinear relationships, and receiver operating characteristic (ROC) curves were used to evaluate diagnostic accuracy. Results A total of 1596 patients with T2DM were included in this study, among whom 736 had OP. After adjusting for covariates, the multivariable logistic regression model showed that compared to participants in the fourth quartile of log2-transformed ALI, those in the first quartile had an increased prevalence of OP in T2DM (OR = 1.95, 95% CI=1.28-2.96, p < 0.01). The multivariable linear regression model indicated that a low log2-transformed ALI is associated with a low femur BMD.RCS demonstrated a linear dose-response relationship between the ALI index and OP in T2DM (p = 0.686), with the area under the ROC curve being 0.57 (95% CI: 0.54-0.60, p < 0.001), and the optimal cutoff value was 6.04. Conclusion Our findings indicate that low levels of ALI are independently associated with an increased prevalence of OP in middle-aged and older adults with T2DM in the United States. ALI may serve as a potential biomarker for assessing the prevalence of OP in middle-aged and older adults with T2DM.
Collapse
Affiliation(s)
- Yifeng Xu
- School of Clinical Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Zhaoqi Yan
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Liangji Liu
- Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| |
Collapse
|
46
|
Wang X, Ni Y, Wang Z, Li C, Hui X, Xu H. Metabolic factors for liver cirrhosis: A Mendelian randomization study. Medicine (Baltimore) 2024; 103:e40507. [PMID: 39809143 PMCID: PMC11596572 DOI: 10.1097/md.0000000000040507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 10/24/2024] [Indexed: 01/16/2025] Open
Abstract
Liver cirrhosis is a chronic disease caused by long-term inflammation and fibrosis of the liver. Early identification and intervention in liver cirrhosis have become an important goal for researchers to explore the influence of some metabolic factors on the risk of liver cirrhosis in terms of genetic susceptibility. Data from genome-wide association studies (GWASs) of fourteen metabolic factors and liver cirrhosis were obtained from publicly available databases. To make the results more credible, we selected 2 GWASs for liver cirrhosis to be validated separately. The causal effect of metabolic factors on liver cirrhosis was assessed separately using 2-sample Mendelian Randomization (MR). The inverse variance weighted (IVW) method was used as the main analysis method. The present MR analysis confirmed that fasting insulin level (IVW-OR = 2.89, 95% CI: 1.36-6.15, P = .006) and ALT (IVW-OR = 1.42, 95% CI: 1.11-1.80, P = .004) were positively causally associated with the risk of liver cirrhosis, and there was a negative causal relationship between hypertension and the risk of liver cirrhosis (IVW-OR = 0.40, 95% CI: 0.23-0.72, P = .002) in 1 liver cirrhosis GWAS. In replication analysis, our MR proved the positive causal effect between ALT (IVW-OR = 2.09, 95% CI: 1.61-2.72, P < .001) and BMI (IVW-OR = 1.44, 95% CI: 1.17-1.77, P < .001) and the risk of liver cirrhosis. A causal relationship between other metabolic factors and the risk of liver cirrhosis could not be established in the current selection of data. Our MR study revealed a causal and positive association between ALT and the risk of liver cirrhosis, suggesting an important role of effective control of ALT in liver cirrhosis prevention. The causal relationship between thirteen other metabolic factors and the risk of liver cirrhosis remains to be further verified.
Collapse
Affiliation(s)
- Xiaobing Wang
- Department of Gastroenterology, The First Affiliated hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yu Ni
- Department of Gastroenterology, The First Affiliated hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Ziwen Wang
- Department of Gastroenterology, The First Affiliated hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Changhui Li
- Department of Gastroenterology, The First Affiliated hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xinyu Hui
- Department of Gastroenterology, The First Affiliated hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Hongyu Xu
- Department of Gastroenterology, The First Affiliated hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
47
|
Choi W, Park M, Park S, Park JY, Hong AR, Yoon JH, Ha KH, Kim DJ, Kim HK, Kang HC. Combined impact of prediabetes and hepatic steatosis on cardiometabolic outcomes in young adults. Cardiovasc Diabetol 2024; 23:422. [PMID: 39574105 PMCID: PMC11583572 DOI: 10.1186/s12933-024-02516-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 11/17/2024] [Indexed: 11/24/2024] Open
Abstract
OBJECTIVES This study aimed to investigate the impact of hepatic steatosis on cardiometabolic outcomes in young adults with prediabetes. METHODS A nationwide cohort study was conducted with 896,585 young adults under 40 years old without diabetes or previous history of cardiovascular disease. Hepatic steatosis was identified using a fatty liver index of ≥ 60. The outcomes of this study were incident diabetes (DM) and composite major adverse cardiovascular events (MACE), including myocardial infarction, stroke, or cardiovascular death. RESULTS During a median follow-up of 11.8 years, 27,437 (3.1%) incident DM cases and 6,584 (0.7%) MACE cases were recorded. Young adults with prediabetes had a significantly higher risk of incident DM (hazard ratio [HR]: 2.81; 95% confidence interval [CI]: 2.74-2.88; P-value: <0.001) and composite MACE risk (HR: 1.10; 95% CI: 1.03-1.17; P-value: 0.003) compared to individuals with normoglycemia, after adjusting for relevant covariates. Stratification based on hepatic steatosis showed that the combination of prediabetes and hepatic steatosis posed the highest risk for these outcomes, after adjusting for relevant covariates. For incident DM, the HRs (95% CI; P-value) were: 3.15 (3.05-3.26; <0.001) for prediabetes without hepatic steatosis, 2.89 (2.78-3.01; <0.001) for normoglycemia with hepatic steatosis, and 6.60 (6.33-6.87; <0.001) for prediabetes with hepatic steatosis. For composite MACE, the HRs (95% CI; P-value) were 1.05 (0.97-1.13; 0.235) for prediabetes without hepatic steatosis, 1.39 (1.27-1.51; <0.001) for normoglycemia with hepatic steatosis, and 1.60 (1.44-1.78; <0.001) for prediabetes with hepatic steatosis. CONCLUSIONS Prediabetes and hepatic steatosis additively increased the risk of cardiometabolic outcomes in young adults. These findings hold significance for physicians as they provide insights into assessing high-risk individuals among young adults with prediabetes.
Collapse
Affiliation(s)
- Wonsuk Choi
- Department of Internal Medicine, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, 322, Seoyang-ro, Hwasun-eup, Hwasun-gun, Hwasun, 58128, Jeollanam-do, Republic of Korea.
- Department of Biological Chemistry, University of California Irvine School of Medicine, Irvine, CA, USA.
| | - Minae Park
- Data Science Team, Hanmi Pharm. Co., Ltd, Seoul, Korea
| | - Sojeong Park
- Data Science Team, Hanmi Pharm. Co., Ltd, Seoul, Korea
| | - Ji Yong Park
- Department of Internal Medicine, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, 322, Seoyang-ro, Hwasun-eup, Hwasun-gun, Hwasun, 58128, Jeollanam-do, Republic of Korea
| | - A Ram Hong
- Department of Internal Medicine, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, 322, Seoyang-ro, Hwasun-eup, Hwasun-gun, Hwasun, 58128, Jeollanam-do, Republic of Korea
| | - Jee Hee Yoon
- Department of Internal Medicine, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, 322, Seoyang-ro, Hwasun-eup, Hwasun-gun, Hwasun, 58128, Jeollanam-do, Republic of Korea
| | - Kyoung Hwa Ha
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon, Korea
| | - Dae Jung Kim
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon, Korea
| | - Hee Kyung Kim
- Department of Internal Medicine, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, 322, Seoyang-ro, Hwasun-eup, Hwasun-gun, Hwasun, 58128, Jeollanam-do, Republic of Korea.
| | - Ho-Cheol Kang
- Department of Internal Medicine, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, 322, Seoyang-ro, Hwasun-eup, Hwasun-gun, Hwasun, 58128, Jeollanam-do, Republic of Korea
| |
Collapse
|
48
|
Besné-Eseverri I, Martín MÁ, Lobo G, Cano MP, Portillo MP, Trepiana J. Antioxidant and Anti-Inflammatory Effects of Opuntia Extracts on a Model of Diet-Induced Steatosis. Antioxidants (Basel) 2024; 13:1416. [PMID: 39594557 PMCID: PMC11591152 DOI: 10.3390/antiox13111416] [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: 10/03/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
Oxidative stress and inflammation are widely recognised as factors that can initiate and facilitate the development of MAFLD. The aim of this study is to analyse the effect of low and high doses of Opuntia stricta var. dillenii peel extract (L-OD and H-OD, respectively) and Opuntia ficus-indica var. colorada pulp extract (L-OFI and H-OFI, respectively), which are rich in betalains and phenolic compounds, on oxidative stress, inflammation, DNA damage and apoptosis in rat livers with diet-induced steatosis. Steatotic diet led to increased final body and liver weight, serum transaminases, hepatic TG content, oxidative status and cell death. H-OFI treatment decreased serum AST levels, while L-OFI reduced hepatic TG accumulation. Oxidative stress was partially prevented with H-OD and H-OFI supplementation, and pro-inflammatory cytokines levels were especially improved with H-OFI treatment. Moreover, H-OFI appears to prevent DNA damage markers. Finally, H-OD and L-OFI supplementation down-regulated the apoptotic pathway. In conclusion, both H-OD and H-OFI supplementation were effective in regulating the progression to metabolic steatohepatitis, triggering different mechanisms of action.
Collapse
Affiliation(s)
- Irene Besné-Eseverri
- Nutrition and Obesity Group, Department of Nutrition and Food Sciences, Faculty of Pharmacy, University of the Basque Country (UPV/EHU) and Lucio Lascaray Research Centre, 01006 Vitoria-Gasteiz, Spain; (I.B.-E.); (M.P.P.)
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), Institute of Health Carlos III, 28029 Madrid, Spain
| | - María Ángeles Martín
- Science and Food Technology and Nutrition Institute (ICTAN-CSIC), 28040 Madrid, Spain;
- CIBER Diabetes and Related Metabolic Diseases (CIBERdem), Institute of Health Carlos III, 28029 Madrid, Spain
| | - Gloria Lobo
- Department of Crop Production in Tropical and Subtropical Areas, Instituto Canario de Investigaciones Agrarias (ICIA), 38297 Tenerife, Spain;
| | - M. Pilar Cano
- Laboratory of Phytochemistry and Plant Food Functionality, Biotechnology and Food Microbiology Department, Institute of Food Science Research (CIAL) (CSIC-UAM), Nicolás Cabrera 9, 28049 Madrid, Spain;
| | - María P. Portillo
- Nutrition and Obesity Group, Department of Nutrition and Food Sciences, Faculty of Pharmacy, University of the Basque Country (UPV/EHU) and Lucio Lascaray Research Centre, 01006 Vitoria-Gasteiz, Spain; (I.B.-E.); (M.P.P.)
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), Institute of Health Carlos III, 28029 Madrid, Spain
- BIOARABA Institute of Health, 01009 Vitoria-Gasteiz, Spain
| | - Jenifer Trepiana
- Nutrition and Obesity Group, Department of Nutrition and Food Sciences, Faculty of Pharmacy, University of the Basque Country (UPV/EHU) and Lucio Lascaray Research Centre, 01006 Vitoria-Gasteiz, Spain; (I.B.-E.); (M.P.P.)
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), Institute of Health Carlos III, 28029 Madrid, Spain
- BIOARABA Institute of Health, 01009 Vitoria-Gasteiz, Spain
| |
Collapse
|
49
|
Brai BIC, Joseph RO, Komolafe TR, Amosun BE, Crown OO, Komolafe K, Ogungbe IV. Neem seed oil ameliorates diabetic phenotype by suppressing redox imbalance, dyslipidaemia and pro-inflammatory mediators in a rodent model of type 2 diabetes. Arch Physiol Biochem 2024:1-15. [PMID: 39548959 DOI: 10.1080/13813455.2024.2426497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 10/02/2024] [Accepted: 10/31/2024] [Indexed: 11/18/2024]
Abstract
The neem plant (Azadirachta indica) has popular ethnomedicinal applications. The anti-diabetic potential and mechanism of neem seed oil (NSO) in a rodent model of type 2 diabetes mellitus was evaluated in the present study. Experimentally-induced diabetic animals were administered NSO (200 and 400 mg/kg) or metformin (150 mg/kg) orally for 30 days, with some animals serving as positive and negative controls. NSO significantly (p < .05) reversed diabetes-induced impaired glucose metabolism, dyslipidaemia, and oxido-inflammatory imbalances typified by changes in the NADH/NAD+ ratio (p < .001) and increases in the mRNA or protein levels of C-reactive protein, 4-hydroxynonenal, and pro-inflammatory cytokines (TNF-α and Il-1β) among others in the hepatic or pancreatic tissues of diabetic animals. The histological evaluation of the pancreatic tissue corroborated the protective effect of NSO. The findings showed that the antidiabetic effect of NSO proceeded through its hypolipidemic effect and modulation of redox and inflammatory signalling events in the tissues of animals.
Collapse
Affiliation(s)
- Bartholomew I C Brai
- Department of Biochemistry, Faculty of Science, Federal University Oye-Ekiti, Oye-Ekiti, Nigeria
| | - Ruth Ometere Joseph
- Department of Biochemistry, Faculty of Science, Federal University Oye-Ekiti, Oye-Ekiti, Nigeria
| | - Titilope Ruth Komolafe
- Department of Biochemistry, Faculty of Science, Federal University Oye-Ekiti, Oye-Ekiti, Nigeria
| | - Busayo Elizabeth Amosun
- Department of Biochemistry, Faculty of Science, Federal University Oye-Ekiti, Oye-Ekiti, Nigeria
| | | | - Kayode Komolafe
- Department of Biochemistry, Faculty of Science, Federal University Oye-Ekiti, Oye-Ekiti, Nigeria
| | | |
Collapse
|
50
|
He C, Zhang Q, Zhu R, Tse G, Wong WT. Asperuloside activates hepatic NRF2 signaling to stimulate mitochondrial metabolism and restore lipid homeostasis in high fat diet-induced MAFLD. Eur J Pharmacol 2024; 983:177003. [PMID: 39278309 DOI: 10.1016/j.ejphar.2024.177003] [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: 07/25/2024] [Revised: 09/03/2024] [Accepted: 09/13/2024] [Indexed: 09/18/2024]
Abstract
BACKGROUND Nutrient overload predisposes the development of metabolic dysfunction-associated fatty liver disease (MAFLD). However, there are no specific pharmacological therapies for MAFLD. Asperuloside (ASP), an iridoid glycoside extracted from Eucommia ulmoides leaves, can alleviate obesity and MAFLD. However, the underlying mechanism and pharmacological effects of ASP on ameliorating MAFLD remain largely investigated. This study aimed to explore the effects of ASP in ameliorating MAFLD and to unravel its underlying mechanism using a high fat diet-induced MAFLD mice model. METHODS Six-week-old C57BL/6 male mice were fed a high fat diet for 12 weeks to induce MAFLD, followed by daily ASP treatment (50 mg/kg via oral gavage) for 7 weeks. HepG2 cells were used for in vitro studies. Nuclear factor erythroid 2-related factor 2 (Nrf2) inhibitor, ML385, was employed to explore the mechanisms of ASP's action. RESULTS ASP stimulated lipolysis and inhibited de novo lipogenesis, contributing to alleviating lipid deposition in obese mice livers and HepG2 cells. ASP restored ATP production and reversed the impairments of mitochondrial energetics and biogenesis in obese mice livers and HepG2 cells. ASP attenuated oxidative stress in obese mice livers and HepG2 cells, exhibiting its antioxidant value. Impressively, ASP significantly promotes Nrf2 nuclear translocation and Nrf2/ARE binding, thereby activating Nrf2/ARE pathway in obese mice livers and HepG2 cells, demonstrating its potential as a hepatic Nrf2 activator. Nrf2 inhibition abolishes the protective effects of ASP against lipid deposition, oxidative stress and mitochondrial dysfunction, emphasizing the critical role of ASP-activated hepatic Nrf2 signaling in ameliorating MAFLD. CONCLUSIONS This study provides the first line of evidence demonstrating the pivotal role of ASP-stimulated Nrf2 activation in alleviating MAFLD, emphasizing its potential as a hepatic Nrf2 activator targeting fatty liver diseases. These findings offer new evidence of ASP-stimulated mitochondrial metabolism and lipolysis in MAFLD, paving the way for the development of ASP as a therapeutic agent and dietary supplement to attenuate MAFLD progression.
Collapse
Affiliation(s)
- Chufeng He
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China; State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong 999077, China.
| | - Qile Zhang
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China; State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Ruiwen Zhu
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China; State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Gary Tse
- School of Nursing and Health Studies, Hong Kong Metropolitan University, 999077, China; Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Wing Tak Wong
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China; Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China; State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong 999077, China.
| |
Collapse
|