1
|
Liu M, Li Y, Li W, Tao Q, Liu F, Zhou J, Yan Z, Zhang X, Yang H, Li S, Wang Z. Hemp seed protein exerts its hypoglycemic and hypolipidemic effects through degradation into short peptides. Food Chem 2025; 484:144406. [PMID: 40279904 DOI: 10.1016/j.foodchem.2025.144406] [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: 11/07/2024] [Revised: 04/05/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025]
Abstract
This study aimed to investigate the effects of hemp seed protein (HSP) on glucose and lipid metabolism and its possible mechanisms. In a high-fat-induced mouse obesity model, HSP supplementation significantly reduced serum TC (Total cholesterol), TG (Triglycerides), and LDL-C (Low-density lipoprotein cholesterol) levels by 28 % (P < 0.001), 34 % (P < 0.001), and 40 % (P < 0.001) respectively, compared to the model group, while HDL-C (High-density lipoprotein cholesterol) increased by 77 % (P < 0.001). Hepatic lipid accumulation was alleviated, and glucose tolerance and insulin sensitivity improved. In vitro, HSP hydrolysates exhibited stronger inhibitory activity against pancreatic α-amylase and lipase than HSP itself. Network pharmacology and molecular docking identified three hemp seed peptides from HSP hydrolysates, which interacted with AKT1, PPARG, and HMGCR. These findings suggest that the metabolic regulatory effects of HSP are mediated by bioactive peptides that inhibit digestive enzymes and regulate AMPK-AKT1/PPARG/HMGCR metabolism pathway, providing insights into its potential as a functional health food.
Collapse
Affiliation(s)
- Mingchen Liu
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China; Chinese Medicinal Materials Breeding Innovation Center, Yuelushan Laboratory, 410128, China.
| | - Yicheng Li
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China; Fine Biotechnological R&D Center, Guangzhou 510000, China.
| | - Wentao Li
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China; Chinese Medicinal Materials Breeding Innovation Center, Yuelushan Laboratory, 410128, China.
| | - Qian Tao
- The Infinitus Global R&D Center, Guangzhou 510000, China.
| | - Fei Liu
- Fine Biotechnological R&D Center, Guangzhou 510000, China.
| | - Jiwei Zhou
- Independent Researcher, Campbell, CA 95008, USA.
| | - Zhiwei Yan
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China; Chinese Medicinal Materials Breeding Innovation Center, Yuelushan Laboratory, 410128, China.
| | - Xiaoyan Zhang
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China
| | - Hua Yang
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China; Chinese Medicinal Materials Breeding Innovation Center, Yuelushan Laboratory, 410128, China
| | - Shuangqi Li
- Fine Biotechnological R&D Center, Guangzhou 510000, China.
| | - Zheng Wang
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China; Chinese Medicinal Materials Breeding Innovation Center, Yuelushan Laboratory, 410128, China.
| |
Collapse
|
2
|
Sadri M, Shafaghat Z, Roozbehani M, Hoseinzadeh A, Mohammadi F, Arab FL, Minaeian S, Fard SR, Faraji F. Effects of Probiotics on Liver Diseases: Current In Vitro and In Vivo Studies. Probiotics Antimicrob Proteins 2025; 17:1688-1710. [PMID: 39739162 DOI: 10.1007/s12602-024-10431-z] [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] [Accepted: 12/06/2024] [Indexed: 01/02/2025]
Abstract
Various types of liver or hepatic diseases cause the death of about 2 million people worldwide every year, of which 1 million die from the complications of cirrhosis and another million from hepatocellular carcinoma and viral hepatitis. Currently, the second most common solid organ transplant is the liver, and the current rate represents less than 10% of global transplant requests. Hence, finding new approaches to treat and prevent liver diseases is essential. In liver diseases, the interaction between the liver, gut, and immune system is crucial, and probiotics positively affect the human microbiota. Probiotics are a non-toxic and biosafe alternative to synthetic chemical compounds. Health promotion by lowering cholesterol levels, stimulating host immunity, the natural gut microbiota, and other functions are some of the activities of probiotics, and their metabolites, including bacteriocins, can exert antimicrobial effects against a broad range of pathogenic bacteria. The present review discusses the available data on the results of preclinical and clinical studies on the effects of probiotic administration on different types of liver diseases.
Collapse
Affiliation(s)
- Maryam Sadri
- Department of Immunology, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Shafaghat
- Department of Immunology, Iran University of Medical Sciences, Tehran, Iran
| | - Mona Roozbehani
- Vaccine Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Akram Hoseinzadeh
- Cancer Research Center, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Fatemeh Mohammadi
- Department of Immunology, School of Medicine, Mashhad University of Medicine Sciences, Mashhad, Iran
| | - Fahimeh Lavi Arab
- Department of Immunology, School of Medicine, Mashhad University of Medicine Sciences, Mashhad, Iran
| | - Sara Minaeian
- Antimicrobial Resistance Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medicine Sciences, Tehran, Iran
| | - Soheil Rahmani Fard
- Antimicrobial Resistance Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medicine Sciences, Tehran, Iran
| | - Fatemeh Faraji
- Antimicrobial Resistance Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medicine Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Felicianna, Lo EKK, Chen C, Ismaiah MJ, Zhang F, Leung HKM, El-Nezami H. Alpha-aminobutyric acid ameliorates diet-induced metabolic dysfunction-associated steatotic liver disease (MASLD) progression in mice via enhancing AMPK/SIRT1 pathway and modulating the gut-liver axis. J Nutr Biochem 2025; 140:109885. [PMID: 40015656 DOI: 10.1016/j.jnutbio.2025.109885] [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: 09/16/2024] [Revised: 12/12/2024] [Accepted: 02/21/2025] [Indexed: 03/01/2025]
Abstract
Alpha-aminobutyric acid (ABA) is a nonproteinogenic amino acid, a metabolite which could be generated from the metabolism of methionine, threonine, serine and glycine or as a gut-microbiome-derived metabolite. Changes in ABA levels have been embroiled in metabolic dysfunction-associated steatotic liver disease (MASLD) intervention studies, but their relation to MASLD pathogenesis remains unclear. Hence, this present study aimed to investigate the effect of oral ABA supplementation on the progression of a high fat/high cholesterol diet (HFD) induced MASLD mice model. ABA was found to remodel the gut microbiome composition and ameliorate MASLD parameters in HFD-fed mice. ABA mitigated HFD-induced gain in liver weight, hepatic steatosis, insulin resistance, serum and hepatic triglyceride levels, and liver cholesterol levels. Modulation of lipid metabolism was observed in the liver, in which expression of proteins and/or genes involved in de novo lipogenesis were suppressed, while those involved in fatty acid oxidation and autophagy were upregulated together with cellular antioxidant capacity, in addition to the enhancement of the AMPK/SIRT1 pathway. ABA reshaped the gut composition by enriching nine bacterial species, including Helicobacter hepaticus, Desulfovibrio sp. G11, Parabacteroides distasonis, and Bacteroides fragilis, while diminishing the abundance of 16 species, which included four Helicobacter species. KEGG pathway analysis of microbial functions found that ABA impeded secondary bile acid biosynthesis - which was reflected in the faecal BA composition analysis. Notably, ABA also inhibited ileal FXR-Fgf15 signaling, allowing for increased hepatic Cyp7a1 expression to eliminate cholesterol buildup in the liver. Overall, our findings indicate that ABA could be used as a promising therapeutic approach for the intervention of MASLD.
Collapse
Affiliation(s)
- Felicianna
- School of Biological Sciences, University of Hong Kong, Pokfulam, Hong Kong, China
| | - Emily Kwun Kwan Lo
- School of Biological Sciences, University of Hong Kong, Pokfulam, Hong Kong, China
| | - Congjia Chen
- School of Biological Sciences, University of Hong Kong, Pokfulam, Hong Kong, China
| | | | - Fangfei Zhang
- School of Biological Sciences, University of Hong Kong, Pokfulam, Hong Kong, China
| | | | - Hani El-Nezami
- School of Biological Sciences, University of Hong Kong, Pokfulam, Hong Kong, China; Institute of Public Health and Clinical Nutrition, School of Medicine, University of Eastern Finland, Kuopio, Finland.
| |
Collapse
|
4
|
Mandal AK, Sahoo A, Almalki WH, Almujri SS, Alhamyani A, Aodah A, Alruwaili NK, Abdul Kadir SZBS, Mandal RK, Almalki RA, Lal JA, Rahman M. Phytoactives for Obesity Management: Integrating Nanomedicine for Its Effective Delivery. Nutr Rev 2025; 83:1152-1170. [PMID: 39331591 DOI: 10.1093/nutrit/nuae136] [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] [Indexed: 09/29/2024] Open
Abstract
Obesity is a global health concern that requires urgent investigation and management. While synthetic anti-obesity medications are available, they come with a high risk of side-effects and variability in their efficacy. Therefore, natural compounds are increasingly being used to treat obesity worldwide. The proposition that naturally occurring compounds, mainly polyphenols, can be effective and safer for obesity management through food and nutrient fortification is strongly supported by extensive experimental research. This review focuses on the pathogenesis of obesity while reviewing the efficacy of an array of phytoactives used for obesity treatment. It details mechanisms such as enzyme inhibition, energy expenditure, appetite suppression, adipocyte differentiation, lipid metabolism, and modulation of gut microbiota. Comprehensive in vitro, in vivo, and preclinical studies underscore the promise of phytoactives in combating obesity, which have been thoroughly reviewed. However, challenges, such as poor bioavailability and metabolism, limit their potential. Advances in nanomedicines may overcome these constraints, offering a new avenue for enhancing the efficacy of phytoactives. Nonetheless, rigorous and targeted clinical trials are essential before applying phytoactives as a primary treatment for obesity.
Collapse
Affiliation(s)
- Ashok Kumar Mandal
- Department of Pharmacology, Faculty of Medicine, University Malaya, Kuala Lumpur 50603, Malaysia
| | - Ankit Sahoo
- Department of Pharmaceutical Sciences, Shalom Institute of Health & Allied Sciences, Sam Higginbottom University of Agriculture, Technology & Sciences, Allahabad, Uttar Pradesh 211007, India
| | - Waleed H Almalki
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Salem Salman Almujri
- Department of Pharmacology, College of Pharmacy, King Khalid University, Asir-Abha 61421, Saudi Arabia
| | - Abdulrahman Alhamyani
- Pharmaceuticals Chemistry Department, Faculty of Clinical Pharmacy, Al Baha University, Al Baha 65779, Saudi Arabia
| | - Alhussain Aodah
- College of Pharmacy, Prince Sattam bin Abdulaziz University, Alkharj 11942, Saudi Arabia
| | - Nabil K Alruwaili
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Sakakah 72341, Saudi Arabia
| | | | | | - Rami A Almalki
- Clinical Pharmacy Unit, Pharmaceutical Care Department, King Faisal Hospital, Makkah Health Cluster, Makkah 24382, Saudi Arabia
| | - Jonathan A Lal
- Department of Molecular and Cellular Engineering, Jacob Institute of Biotechnology and Bioengineering, Sam Higginbottom University of Agriculture, Technology, and Sciences, Prayagraj, Uttar Pradesh 211007, India
| | - Mahfoozur Rahman
- Department of Pharmaceutical Sciences, Shalom Institute of Health & Allied Sciences, Sam Higginbottom University of Agriculture, Technology & Sciences, Allahabad, Uttar Pradesh 211007, India
| |
Collapse
|
5
|
Hu Y, Sang N, Wu A, Pu J, Yan H, Luo J, Zheng P, Luo Y, Yu J, He J, Yu B, Chen D. Different types of bile acids exhibit opposite regulatory effects on lipid metabolism in finishing pigs through bile acid receptors. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2025; 21:25-36. [PMID: 40135169 PMCID: PMC11930731 DOI: 10.1016/j.aninu.2024.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 11/20/2024] [Accepted: 12/16/2024] [Indexed: 03/27/2025]
Abstract
The purpose of this research was to investigate how different bile acids impact lipid metabolism and carcass characteristics in finishing pigs, along with the potential mechanisms involved. Twenty-one finishing pigs (Duroc×Landrace×Yorkshire [DLY]; average BW = 144.38 ± 8.92 kg) were assigned to three dietary treatments, with each treatment containing seven replicates, each consisting of one pig. The three dietary treatments included: a basic diet, a basic diet supplemented with 500 mg/kg of hyodeoxycholic acid (HDCA), and a basic diet supplemented with 500 mg/kg of lithocholic acid (LCA). The trial lasted for 28 d. Hyodeoxycholic acid was used in the in vitro experiments and added to mature 3T3-L1 adipocytes for 4 d to elucidate the mechanism by which bile acids regulate lipid metabolism. The results suggested that HDCA tended to decrease backfat thickness in finishing pigs (P = 0.094) and reduced the size of lipid droplets in 3T3-L1 adipocytes (P = 0.012), whereas LCA increased backfat thickness (P = 0.016) and induced larger lipid droplets in the abdominal adipose tissue (P = 0.003). Furthermore, HDCA enhanced the expression of Takeda G-protein-coupled receptor 5 protein and hormone-sensitive lipase (HSL) gene in backfat of pigs (P < 0.05) and increased the protein expression of phosphorylated HSL (p-HSL) in vitro (P = 0.093). Compared to HDCA, LCA addition increased the gene and protein expression of peroxisome proliferator activated receptor gamma in backfat of pigs (P < 0.05) and enhanced the expression of hepatic genes sterol regulatory element-binding protein-1c and fatty acid synthase (P < 0.05). In conclusion, HDCA enhanced lipolysis and partially decreased backfat thickness in finishing pigs, while LCA promoted lipid synthesis and increased backfat thickness of pigs. The variations in the effects of various bile acids on bile acid receptors could explain these functional differences.
Collapse
Affiliation(s)
- Yaolian Hu
- Key Laboratory of Animal Disease-resistant Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Ya'an 625014, China
| | - Ni Sang
- Key Laboratory of Animal Disease-resistant Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Ya'an 625014, China
| | - Aimin Wu
- Key Laboratory of Animal Disease-resistant Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Ya'an 625014, China
| | - Junning Pu
- Key Laboratory of Animal Disease-resistant Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Ya'an 625014, China
| | - Hui Yan
- Key Laboratory of Animal Disease-resistant Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Ya'an 625014, China
| | - Junqiu Luo
- Key Laboratory of Animal Disease-resistant Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Ya'an 625014, China
| | - Ping Zheng
- Key Laboratory of Animal Disease-resistant Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Ya'an 625014, China
| | - Yuheng Luo
- Key Laboratory of Animal Disease-resistant Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Ya'an 625014, China
| | - Jie Yu
- Key Laboratory of Animal Disease-resistant Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Ya'an 625014, China
| | - Jun He
- Key Laboratory of Animal Disease-resistant Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Ya'an 625014, China
| | - Bing Yu
- Key Laboratory of Animal Disease-resistant Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Ya'an 625014, China
| | - Daiwen Chen
- Key Laboratory of Animal Disease-resistant Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Ya'an 625014, China
| |
Collapse
|
6
|
Twum E, Ofosu-Boateng M, Nnamani DO, Gebreyesus LH, Yadak N, Kharbanda KK, Gonzalez FJ, Gyamfi MA. Blockade of the estrogen receptor alpha-pregnane X receptor axis protects ovariectomized mice against ethanol-induced hepatotoxicity. J Biol Chem 2025:110238. [PMID: 40381698 DOI: 10.1016/j.jbc.2025.110238] [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: 12/02/2024] [Revised: 04/29/2025] [Accepted: 05/05/2025] [Indexed: 05/20/2025] Open
Abstract
Women develop alcohol-associated liver disease (ALD) faster than men at any level of alcohol consumption, implicating estrogen as a contributing factor. However, the precise mechanism remains unknown. Therefore, 12-weeks-old female C57BL/6N mice were subjected to either bilateral ovariectomy (OVX) or sham surgery. After a three-week recovery period, the mice were fed either a 5% ethanol (EtOH)-containing liquid diet or paired-fed control diet for 10 days followed by a single gavage dose of EtOH (5 g/kg, 30% EtOH solution). The mice were examined for serum biochemical parameters, hepatotoxicity, histology, expression of xenobiotic nuclear receptors PXR and CAR, and their target gene mRNAs and proteins in hepatic and perigonadal white adipose tissues (pgWAT). While OVX mice on a control diet significantly gained weight, EtOH significantly increased hepatotoxicity, residual EtOH levels, lipid peroxidation, and oxidative stress in sham-operated mice but not in their OVX counterparts. Additionally, in the livers and pgWAT of the sham mice, EtOH significantly increased the mRNA and/or protein levels of the major estrogen receptor ERα, PXR, CAR, and their target genes, proinflammatory cytokines and chemokines, lipogenic genes, and FGF21 levels, a predictive biomarker for ALD severity in humans, but inhibited NRF2 and its targets genes encoding NQO1 and BHMT. Unexpectedly, all these changes were attenuated in the EtOH-fed OVX mice by the upregulation of NRF2 and aryl hydrocarbon receptor (AhR) and their downstream antioxidant target genes. Together these results suggest the existence of an estrogen-regulated ERα-PXR-NRF2-signaling axis in liver and pgWAT which contributes to sexual dimorphism in ALD.
Collapse
Affiliation(s)
- Elizabeth Twum
- Department of Pharmaceutical Sciences, The University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN, 38163, USA
| | - Malvin Ofosu-Boateng
- Department of Pharmaceutical Sciences, The University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN, 38163, USA
| | - Daniel O Nnamani
- Department of Pharmaceutical Sciences, The University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN, 38163, USA
| | - Lidya H Gebreyesus
- Department of Pharmaceutical Sciences, The University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN, 38163, USA
| | - Nour Yadak
- Department of Pathology and Laboratory Medicine, The University of Tennessee Health Science Center, Memphis, TN, 38163
| | - Kusum K Kharbanda
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, 68105 USA; Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA; Department of Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Frank J Gonzalez
- Center for Cancer Research, National Cancer Institute, Building 37, Room 3106, Bethesda, MD, 20892, USA
| | - Maxwell A Gyamfi
- Department of Pharmaceutical Sciences, The University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN, 38163, USA.
| |
Collapse
|
7
|
Urlandini L, Leonetti AE, Conforti F, Perri A, Lofaro D, Antonucci G, Mandalà M, Bossio S, Di Agostino S, Rago V. Calorie-restriction treatment mitigates the aging in rat liver model. Biogerontology 2025; 26:108. [PMID: 40332584 PMCID: PMC12058891 DOI: 10.1007/s10522-025-10245-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Accepted: 04/22/2025] [Indexed: 05/08/2025]
Abstract
The aging process promotes progressive impairment of homeostasis and the increase of the risk of disease and death. A major hallmark of the aging process is the systemic chronic inflammation which strongly contributes to the onset of aging-related diseases. In the liver, the aging condition drives the hepatocytes to develop a metabolic dysfunction-associated steatosis. Caloric restriction (CR) is a remarkable strategy to delay biological aging, occurring through several mechanisms. In this study we aimed to explore, employing an in vivo rat model, the impact of CR on aging-mediated liver inflammation markers. The experiments were performed on 14 male Sprague-Dawley rats (24 months old). At 18 months old, rats were allocated into two groups: the normal diet (ND) group was continued ad libitum diet, and the CR regimen group was fed a diet of the same chow restricted to 60% of the intake. All animals were sacrificed at 24 months old. Compared to the ND group, morphological examination of the liver revealed a lower level of fibrosis in the CR group, concomitantly with a reduced expression of key fibrotic markers, such as collagen I, fibronectin, and αSMA. Furthermore, CR improved the liver oxidative balance, as showed by the increased expression of two scavenging enzymes, SOD1/SOD. Moreover, we reported concomitant reduction of NLRP3 inflammasome signalling. Interestingly, CR significantly improved the signalling of key members of the nutrition-sensitizing affected by aging, AMPK/SIRT1/LKB1. Collectively our findings support the evidence on the metabolic benefits of CR about aging-related liver inflammation, by inducing a morphological improvement that mirrors the decrease in the expression of inflammatory molecular markers.
Collapse
Affiliation(s)
- L Urlandini
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - A E Leonetti
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - F Conforti
- Pathology Unit, Annunziata Hospital, 87100, Cosenza, Italy
| | - A Perri
- Department of Experimental and Clinical Medicine, Magna Græcia University of Catanzaro, Catanzaro, Italy
| | - D Lofaro
- University of Calabria, Department of Mathematics and Computer Science, Rende, 87036, Cosenza, Italy
| | - G Antonucci
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - M Mandalà
- Department of Biology, Ecology and Earth Science, University of Calabria, Rende, Italy
| | - S Bossio
- Department of Experimental and Clinical Medicine, Magna Græcia University of Catanzaro, Catanzaro, Italy
| | - S Di Agostino
- Department of Health Sciences, Magna Græcia University of Catanzaro, Catanzaro, Italy.
| | - V Rago
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy.
| |
Collapse
|
8
|
Su Q, Pan H, Hong P, You Y, Wu Y, Zou J, Sun J, Rao G, Liao J, Tang Z, Hu L. Protective effect of curcumin against endoplasmic reticulum stress and lipid metabolism disorders in AFB1-intoxicated duck liver. Mycotoxin Res 2025; 41:359-372. [PMID: 40085329 DOI: 10.1007/s12550-025-00586-1] [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: 12/03/2024] [Revised: 02/27/2025] [Accepted: 03/05/2025] [Indexed: 03/16/2025]
Abstract
Aflatoxin B1 (AFB1) is a stable and highly toxic toxin that causes multi-organ toxicity with sustained ingestion, most typically in the duck liver. Previous research has shown that AFB1 can bring about endoplasmic reticulum stress (ERS) in animals, and ERS is strongly associated with lipid metabolism. However, the relationship between AFB1-induced duck liver toxicity and ERS and lipid metabolism is currently unclear. Great attention has been paid to the prevention and treatment of AFB1 because of its great harm. Curcumin, a natural polyphenol, is notable for its powerful anti-inflammatory and antioxidant properties. Studies have shown curcumin to be protective against afb1-induced avian multi-organ toxicity. However, the effects of curcumin on the liver of ducks exposed to AFB1 are largely unknown. In the present study, we aimed to investigate whether AFB1 exposure induces ERS and lipid metabolism disorders in duck liver, while exploring the positive role of curcumin in it. One-day-old ducks (n = 80) were randomly divided in four groups: control group, AFB1 group (0.1 mg / kg.bw AFB1), Cur group (400 mg/kg curcumin), and AFB1 + Cur group (0.1 mg/kg.bw AFB1 + 400 mg/kg curcumin), and blood and liver were collected for the study after 21 days of continuous administration. Our research has found that AFB1 exposure significantly increases the levels of liver function indicators ALP, AST, and ALT in ducks' serum (P < 0.05). Duck liver undergoes fatty degeneration under the influence of AFB1. Under the effect of curcumin, AFB1-induced structural damage in duck liver was somewhat controlled. Further experimental results showed that AFB1 treatment significantly increased the expression of glucose-regulated protein 78 (P < 0.001), and activated the endoplasmic reticulum stress pathway. Meanwhile, AFB1 inhibited the LKB1-AMPK signaling pathway and disrupted lipid metabolic homeostasis. And curcumin treatment effectively reversed these changes. Overall, our results suggest that curcumin attenuates AFB1-induced hepatotoxicity in ducks by inhibiting ERS and lipid metabolism disorders.
Collapse
Affiliation(s)
- Qian Su
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Hang Pan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Panjing Hong
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Yanli You
- College of Life Science, Yantai University, Yantai City, 264005, Shandong Province, China
| | - Yuhan Wu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Junbo Zou
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Jingping Sun
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Gan Rao
- Guangzhou General Pharmaceutical Research Institute Co., Ltd, Guangzhou, China
| | - Jianzhao Liao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Zhaoxin Tang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Lianmei Hu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
9
|
Wang Y, Sanghvi G, Ballal S, Sharma R, Pathak PK, Shankhyan A, Sun J, Chen Q, Ma Y, Huang L, Liu Y. Molecular mechanisms of lncRNA NEAT1 in the pathogenesis of liver-related diseases, with special focus on therapeutic approaches. Pathol Res Pract 2025; 269:155867. [PMID: 40054160 DOI: 10.1016/j.prp.2025.155867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 01/13/2025] [Accepted: 02/25/2025] [Indexed: 04/19/2025]
Abstract
Liver diseases are a major worldwide health concern, with high rates of dysfunction and mortality. In recent years, a variety of lncRNAs have been studied and discovered to be engaged in numerous cellular-level regulatory mechanisms as competing endogenous RNAs (ceRNAs), which play a significant role in the development of liver-related diseases. A class of RNA molecules known as lncRNAs, which are over 200 nucleotides long, do not translate into proteins. Nuclear Enriched Abundant Transcript 1 (NEAT1) is a type of lncRNA that has a critical function in paraspeckles formation and stability. NEAT1 levels are consistently found to be higher than normal in a number of different types of diseases, as well as patients who have high levels of NEAT1 expression often have a poor prognosis. The significance and mode of action of NEAT1 in liver illnesses, such as nonalcoholic fatty liver disease (NAFLD), alcohol-related liver disease (ALD), liver fibrosis/cirrhosis, hepatocellular carcinoma (HCC), viral hepatitis, and liver injury, are becoming more widely known. In this review, we highlighted significant recent studies concerning the various roles of lncRNA NEAT1 in hepatic diseases. As well as, we reviewed novel therapeutic potential of lncRNAs in several liver-related diseases.
Collapse
Affiliation(s)
- Yahui Wang
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China.
| | - Gaurav Sanghvi
- Marwadi University Research Center, Department of Microbiology, Faculty of Science Marwadi University, Rajkot, Gujarat 360003, India
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Rsk Sharma
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh 531162, India
| | - Piyus Kumar Pathak
- Department of Applied Sciences-Chemistry, NIMS Institute of Engineering & Technology, NIMS University Rajasthan, Jaipur, India
| | - Aman Shankhyan
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab 140401, India
| | - Jiaxuan Sun
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, First Hospital of Jilin University, Changchun 130061, China
| | - Qingmin Chen
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, First Hospital of Jilin University, Changchun 130061, China
| | - Yu Ma
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, First Hospital of Jilin University, Changchun 130061, China
| | - Lei Huang
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, First Hospital of Jilin University, Changchun 130061, China
| | - Yahui Liu
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, First Hospital of Jilin University, Changchun 130061, China.
| |
Collapse
|
10
|
Cho W, Choi SW, Lim DS, Gwon HJ, Abd El-Aty AM, Ahmet Aydemir H, Hong SA, Jeong JH, Jung TW. Donepezil alleviates hepatic steatosis by mitigating ER stress via the AMPK/autophagy pathway. Mol Cell Endocrinol 2025; 601:112523. [PMID: 40118333 DOI: 10.1016/j.mce.2025.112523] [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: 12/30/2024] [Revised: 02/18/2025] [Accepted: 03/15/2025] [Indexed: 03/23/2025]
Abstract
Donepezil (Do), a drug known for its ability to reduce neuronal inflammation and for its use in the treatment of Alzheimer's disease, has shown promise in combating hepatic lipid accumulation in hyperlipidemic conditions and endoplasmic reticulum (ER) stress, a factor associated with alterations in hepatic lipid metabolism. However, the mechanisms by which these problems are alleviated have not been fully elucidated. In this study, we investigated the effects of Do on hepatic lipid metabolism through both in vitro and in vivo studies. We examined the expression of proteins associated with lipogenesis and ER stress via immunoblot analysis, and hepatic lipid accumulation was assessed via oil red O staining. In addition, autophagosome formation was analyzed by counting MDC-positive cells. Our results demonstrated that Do treatment improved hepatic lipid metabolism and reduced the expression of ER stress markers, resulting in decreased lipogenic lipid deposition and apoptosis in the hepatocytes and livers of hyperlipidemic mice. Mechanistically, knocking down AMPK or inhibiting autophagy with 3-methyladenine (3 MA) attenuated the effects of Do on palmitate-exposed hepatocytes. These results suggest that Do alleviates hepatic ER stress via the AMPK/autophagy pathway and AMPK-mediated fatty acid oxidation, resulting in improved hepatic lipid metabolism and reduced hepatic steatosis and apoptosis. Our study provides evidence that Do may be a promising therapeutic approach for Alzheimer's disease patients with metabolic dysfunction-associated steatotic liver disease (MASLD).
Collapse
Affiliation(s)
- Wonjun Cho
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - Sung Woo Choi
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - Do Su Lim
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea; Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Seoul, Republic of Korea
| | - Hyeon Ji Gwon
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea; Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Seoul, Republic of Korea
| | - A M Abd El-Aty
- Department of Pharmacology, Faculty of Veterinary Medicine, Cairo University, 12211, Giza, Egypt; Department of Medical Pharmacology, Medical Faculty, Ataturk University, Erzurum 25240, Turkey.
| | - Hacı Ahmet Aydemir
- Department of Family Medicine, Erzurum Regional Training and Research Hospital, Erzurum 25000, Turkey; Dr. Filiz Dolunay Family Health Center Unit Number:59, Yakutiye, Erzurum, Turkey
| | - Soon Auck Hong
- Department of Pathology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - Ji Hoon Jeong
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea; Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Seoul, Republic of Korea
| | - Tae Woo Jung
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea.
| |
Collapse
|
11
|
Li Z, Zheng G, Fang C, Mei J, Liang H, Yang L. Comparation of brain-targeting chitosan/sodium tripolyphosphate and ovalbumin/sodium carboxymethylcellulose nanoparticles on dihydromyricetin delivery and cognitive impairment in obesity-related Alzheimer's disease. Int J Biol Macromol 2025; 306:141517. [PMID: 40020826 DOI: 10.1016/j.ijbiomac.2025.141517] [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: 11/29/2024] [Revised: 02/02/2025] [Accepted: 02/25/2025] [Indexed: 03/03/2025]
Abstract
The brain-gut axis plays an important role in regulating cognitive ability in obesity-related Alzheimer's disease (AD). In this study, we aimed to investigate the correlation between the barrier penetration ability of the DMY nanodelivery system in vivo and the regulation of the gut-brain axis to alleviate cognitive impairment. Brain-targeted peptide (TGN: TGNYKALHPHNG) and DMY loaded chitosan (CS)/sodium tripolyphosphate (TPP) nanoparticles (TGN-DMY-CS/TPP-NPs) and ovalbumin (OVA)/sodium carboxymethylcellulose (CMC) nanoparticles (TGN-DMY-OVA/CMC-NPs) were prepared. TGN-DMY-CS/TPP-NPs demonstrated superior mucus penetration and BBB targeting ability compared to TGN-DMY-OVA/CMC-NPs, while the latter showed notable intestinal accumulation. TGN-DMY-CS/TPP-NPs treatment significantly increased the relative abundance of Alistipes and Rikenellaceae_RC9_gut_group, and TGN-DMY-OVA/CMC-NPs treatment obviously enhanced the relative abundance of Lactobacillus. Furthermore, both nanoparticles alleviated lipid metabolism disorder, oxidative stress, and inflammation in the liver, reduced oxidative stress and neuroinflammation in the brain, inhibited neuronal apoptosis, and enhanced mitochondrial biogenesis and synaptic plasticity in obesity-related AD mice. Despite different mucus penetration and biodistribution, their similar efficacy in improving obesity-related AD is attributed to the gut-brain bidirectional connection.
Collapse
Affiliation(s)
- Zhiwei Li
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, China; Jiangxi Key Laboratory of Natural Product and Functional Food, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China
| | - Guodong Zheng
- Jiangxi Key Laboratory of Natural Product and Functional Food, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China
| | - Chaoping Fang
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, China
| | - Jingtao Mei
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, China
| | - Hanji Liang
- Jiangxi Key Laboratory of Natural Product and Functional Food, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China
| | - Licong Yang
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, China.
| |
Collapse
|
12
|
Zhou M, Zhang Y, Song Z, Tang S, Liu Z, Pang M, Zhang D, Wu X, Yu X, Wang P, Wei Y. Enhanced bioactivity of honeysuckle-Cassia seeds extracts through Lactobacillus acidophilus and Bacillus subtilis co-fermentation: Impact on alcoholic liver disease and gut microbiota. Food Chem 2025; 486:144463. [PMID: 40339419 DOI: 10.1016/j.foodchem.2025.144463] [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: 12/27/2024] [Revised: 04/13/2025] [Accepted: 04/20/2025] [Indexed: 05/10/2025]
Abstract
This study investigated the hepatoprotective potential of Honeysuckle-Cassia seeds extracts co-fermented with Lactobacillus acidophilus and Bacillus subtilis against alcoholic liver disease (ALD) and gut microbiota dysbiosis. Through network pharmacology analysis, 209 overlapping targets between Honeysuckle-Cassia seeds bioactive components and ALD-related targets were identified, with 39 core targets subsequently determined. Comparative analysis of aqueous extract (AE), Lactobacillus acidophilus fermentation broth (LAF), and mixed bacteria fermentation broth (MBF) revealed that MBF significantly enhanced the content of bioactive compounds: total polysaccharides (72.6 ± 3.8 mg/g), flavonoids (34.7 ± 2.5 mg/g), and saponins (15.2 ± 1.1 mg/g), representing 275 %, 72 %, and 62 % increases over AE, respectively (p < 0.05). In a murine ALD model, MBF intervention (12.5 mL/kg, 30 days) significantly reduced serum markers of liver injury (ALT: 35 %, AST: 28 %, TC: 42 %, TG: 39 %; (p < 0.05) and hepatic oxidative stress (MDA ↓52 %, GSH ↑156.55 %, SOD ↑76.71 %). Mechanistically, MBF suppressed pro-inflammatory cytokines (IL-1β, IL-6, TNF-α) by 40-50 % while elevating anti-inflammatory mediators (IL-4, IL-10, PGE2) 1.6-2.0-fold via AMPK/ACC/SREBP1c signaling modulation. Gut microbiota analysis revealed that MBF restored α-diversity indices (Shannon ↑10.06 %, ACE ↑32.34 %) and reversed alcohol-induced dysbiosis by enriching Lachnospiraceae and Blautia while suppressing Alloprevotella. Structural degradation of plant residues (SEM) confirmed microbial synergy in releasing insoluble-bound phytochemicals (100-400 m/z range). These findings validate co-fermentation as a potent strategy to amplify the hepatoprotective and microbiota-modulating activities of traditional herbs, offering a scientific foundation for developing functional foods against ALD.
Collapse
Affiliation(s)
- Mengtian Zhou
- Key Food Science and Engineering College, Beijing University of Agriculture, Beijing 102206, China
| | - Yingao Zhang
- Key Food Science and Engineering College, Beijing University of Agriculture, Beijing 102206, China
| | - Zeyi Song
- College of Bioscience and Resources Environment, Beijing University of Agriculture, Beijing 102206, China
| | - Shuping Tang
- Key Food Science and Engineering College, Beijing University of Agriculture, Beijing 102206, China
| | - Zhaoxue Liu
- Key Food Science and Engineering College, Beijing University of Agriculture, Beijing 102206, China
| | - Meixia Pang
- Key Food Science and Engineering College, Beijing University of Agriculture, Beijing 102206, China
| | - Di Zhang
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, China
| | - Xiaoyan Wu
- Ningbo Yufangtang Biotechnology Co., Ltd., Ningbo 315012, China
| | - Xindi Yu
- Zhejiang Dayidemei Biotechnology Co., Ltd., Ningbo 315012, China
| | - Pengze Wang
- Ningbo Yufangtang Biotechnology Co., Ltd., Ningbo 315012, China
| | - Ying Wei
- Key Food Science and Engineering College, Beijing University of Agriculture, Beijing 102206, China
| |
Collapse
|
13
|
Chadha P, Aghara H, Johnson D, Sharma D, Odedara M, Patel M, Kumar H, Thiruvenkatam V, Mandal P. Gardenin A alleviates alcohol-induced oxidative stress and inflammation in HepG2 and Caco2 cells via AMPK/Nrf2 pathway. Bioorg Chem 2025; 161:108543. [PMID: 40318507 DOI: 10.1016/j.bioorg.2025.108543] [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: 01/29/2025] [Revised: 04/23/2025] [Accepted: 04/27/2025] [Indexed: 05/07/2025]
Abstract
Chronic alcohol consumption triggers immune responses that lead to cell damage, contributing to alcohol-associated liver disease (ALD). Despite its prevalence, no FDA-approved treatment for ALD currently exists. This study explores the cytoprotective effects of Gardenin A (GarA), a polymethoxylated flavone, for protection against alcohol-induced oxidative stress and inflammation in HepG2 and Caco2 cell lines. GarA was isolated, characterized and, tested in-vitro, showing maximum cell viability at 10 μg/ml using MTT assays. Further, lipid accumulation assay, reactive oxygen species (ROS) estimation and nuclear morphology visualization was carried out using different staining techniques. RT-qPCR was employed to examine the expression of various pro- and anti-inflammatory cytokines, along with Cytochrome P4502E1 (CYP2E1) and Sterol regulatory element binding protein-2 (SREBP2) and tight junction genes crucial for gut barrier integrity. Moreover, ELISA was carried out for key protein targets such as AMPK (phosphorylated and total), TNFα, C5aR1, HO-1 and Nrf2. GarA caused a marked decrease in lipid droplets, ROS levels, and expression of pro-inflammatory cytokines. It showed anti-inflammatory and anti-oxidant activity and helped maintain the gut barrier and nuclear integrity. In-silico studies showed the conserved amino acid interaction and affinity of GarA with C5aR1, and TNFα, compared to the interactions with known inhibitors/activators, further corroborating the results. This study is the first to explore the effects of GarA on ALD, underscoring its potential as an anti-inflammatory and anti-oxidant agent targeting the AMPK/Nrf2 signaling pathway, suggesting its future as a promising therapeutic candidate for mitigating alcohol-induced liver and gut damage.
Collapse
Affiliation(s)
- Prashsti Chadha
- Department of Biological Sciences, P D Patel Institute of Applied Sciences (PDPIAS), Charotar University of Science and Technology, Changa, Anand 388421, Gujarat, India
| | - Hiral Aghara
- Department of Biological Sciences, P D Patel Institute of Applied Sciences (PDPIAS), Charotar University of Science and Technology, Changa, Anand 388421, Gujarat, India
| | - Delna Johnson
- Department of Chemistry, Indian Institute of Technology Gandhinagar, Palaj, 382355, Gujarat, India
| | - Dhrubjyoti Sharma
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, 382355, Gujarat, India
| | - Mitalben Odedara
- Department of Medical Laboratory Technology, Bapubhai Desaibhai Patel Institute of Paramedical Sciences (BDIPS), Charotar University of Science and Technology, Changa, Anand 388421, Gujarat, India
| | - Manali Patel
- Department of Biological Sciences, P D Patel Institute of Applied Sciences (PDPIAS), Charotar University of Science and Technology, Changa, Anand 388421, Gujarat, India
| | - Hemant Kumar
- Department of Medical Laboratory Technology, Bapubhai Desaibhai Patel Institute of Paramedical Sciences (BDIPS), Charotar University of Science and Technology, Changa, Anand 388421, Gujarat, India
| | - Vijay Thiruvenkatam
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, 382355, Gujarat, India
| | - Palash Mandal
- Department of Biological Sciences, P D Patel Institute of Applied Sciences (PDPIAS), Charotar University of Science and Technology, Changa, Anand 388421, Gujarat, India.
| |
Collapse
|
14
|
Zhu Y, Jia Y, Zhang E. Oxidative stress modulation in alcohol-related liver disease: From chinese botanical drugs to exercise-based interventions. Front Pharmacol 2025; 16:1516603. [PMID: 40351443 PMCID: PMC12062749 DOI: 10.3389/fphar.2025.1516603] [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: 10/24/2024] [Accepted: 03/10/2025] [Indexed: 05/14/2025] Open
Abstract
Alcohol-related liver disease (ALD) is a chronic liver injury caused by long-term excessive alcohol consumption, with complex and multifaceted pathological mechanisms. Research indicates that oxidative stress (OS), inflammatory responses, and lipid metabolic disturbances induced by alcohol and its metabolites are primary contributors to hepatocyte injury, positioning OS as a key target in ALD treatment. The main non-pharmacological treatment for ALD is alcohol abstinence, while medical treatment primarily relies on Western pharmacological interventions. However, recent research has increasingly highlighted the potential of Chinese botanical drugs in improving histological features and modulating signaling pathways associated with OS in ALD, underscoring the therapeutic potential of traditional Chinese herb medicine. Despite these promising findings, the precise mechanisms and effects of these extracts remain incompletely understood, and potential side effects must be considered. Therefore, a comprehensive analysis of herbal extracts with therapeutic effects is essential to optimize clinical administration and ensure safe, effective treatment. This review focuses on OS as a central theme, categorizing Chinese botanical drugs into six major groups-flavonoids, polyphenols, terpenoids, alkaloids, saponins, and anthraquinones-all widely used in traditional Chinese herb medicine. The review provides an overview of their botanical characteristics and therapeutic actions in the context of ALD, offering insights into OS regulation and exploring their potential as treatments for ALD. Notably, physical exercise shares overlapping mechanisms with botanical drugs in regulating OS. Combining two strategies could offer a promising integrative treatment for ALD, though further research is needed to confirm their synergistic benefits and optimize clinical applications.
Collapse
Affiliation(s)
| | | | - Enming Zhang
- School of Sports Medicine and Physical Therapy, Beijing Sport University, Beijing, China
| |
Collapse
|
15
|
Zhang F, Yue K, Sun S, Lu S, Jia G, Zha Y, Zhang S, Chou CJ, Liao C, Li X, Duan Y. Targeting Histone Deacetylase 11 with a Highly Selective Inhibitor for the Treatment of MASLD. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412903. [PMID: 39976110 PMCID: PMC12005767 DOI: 10.1002/advs.202412903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 01/26/2025] [Indexed: 02/21/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) represents the most prevalent chronic liver disorder globally. Due to its intricate pathogenesis and the current lack of efficacious pharmacological interventions, there is a pressing need to discover novel therapeutic targets and agents for MASLD treatment. Herein, it is found that histone deacetylase 11 (HDAC11), a subtype of HDAC family, is markedly overexpressed in both in vitro and in vivo models of MASLD. Furthermore, the knockdown of HDAC11 is observed to mitigate lipid accumulation in hepatic cells. A highly selective HDAC11 inhibitor, B6, which exhibits favorable pharmacokinetic property and liver distribution, is further designed and synthesized. Integrating RNA-seq data with in vivo and in vitro experiments, B6 is found to inhibit de novo lipogenesis (DNL) and promote fatty acid oxidation, thus mitigating hepatic lipid accumulation and pathological symptoms in MASLD mice. Further omics analysis and experiments reveal that B6 enhances the phosphorylation of AMPKα1 at Thr172 through the inhibition of HDAC11, consequently modulating DNL and fatty acid oxidation in the liver. In summary, this study identifies HDAC11 as a potential therapeutic target in MASLD and reports the discovery of a highly selective HDAC11 inhibitor with favorable drug-like properties for the treatment of MASLD.
Collapse
Affiliation(s)
- Feng Zhang
- Department of Cardiologythe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefei230001China
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education InstitutesHefei University of TechnologyHefei230031China
| | - Kairui Yue
- Key Laboratory of Marine DrugsChinese Ministry of EducationSchool of Medicine and PharmacyOcean University of China5 Yushan RoadQingdao266003China
| | - Simin Sun
- Key Laboratory of Marine DrugsChinese Ministry of EducationSchool of Medicine and PharmacyOcean University of China5 Yushan RoadQingdao266003China
| | - Shengyuan Lu
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education InstitutesHefei University of TechnologyHefei230031China
| | - Geng Jia
- Key Laboratory of Marine DrugsChinese Ministry of EducationSchool of Medicine and PharmacyOcean University of China5 Yushan RoadQingdao266003China
| | - Yang Zha
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education InstitutesHefei University of TechnologyHefei230031China
| | - Shuang Zhang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education InstitutesHefei University of TechnologyHefei230031China
| | - C. James Chou
- Department of Drug Discovery and Biomedical SciencesCollege of PharmacyMedical University of South CarolinaCharlestonSC29425USA
| | - Chenzhong Liao
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education InstitutesHefei University of TechnologyHefei230031China
| | - Xiaoyang Li
- Key Laboratory of Marine DrugsChinese Ministry of EducationSchool of Medicine and PharmacyOcean University of China5 Yushan RoadQingdao266003China
- Marine Biomedical Research Institute of QingdaoQingdaoShandong266071China
| | - Yajun Duan
- Department of Cardiologythe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefei230001China
| |
Collapse
|
16
|
Barbier-Torres L, Luque-Urbano M, Chhimwal J, Robinson AE, Fernández-Ramos D, Lopitz-Otsoa F, Van Eyk JE, Millet O, Mato JM, Lu SC. Fructose-induced progression of steatohepatitis involves disrupting aldolase B-AMPK signaling in methionine adenosyltransferase 1A deficient mice. Metabolism 2025; 165:156154. [PMID: 39922455 PMCID: PMC12036799 DOI: 10.1016/j.metabol.2025.156154] [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: 10/25/2024] [Revised: 01/23/2025] [Accepted: 02/03/2025] [Indexed: 02/10/2025]
Abstract
OBJECTIVE Aldolases (ALDO) are sensors that regulate AMPK via binding to fructose 1,6-biphosphate (FBP), an intermediate of glucose and fructose metabolism. Fructose consumption is linked to metabolic dysfunction-associated steatotic liver disease (MASLD) progression but whether ALDO-AMPK signaling is involved is unknown. Methionine adenosyltransferase alpha 1 (Mat1a) knockout (KO) mice have low hepatic S-adenosylmethionine (SAMe) level and spontaneously develop steatohepatitis. ALDOB methylation has not been reported and here we investigated whether SAMe level regulates ALDOB and ALDOB-AMPK signaling and whether fructose feeding accelerates MASLD progression by disrupting ALDOB-AMPK signaling. METHODS Mass spectrometry identified ALDOB methylation sites and recombinant in vitro approaches assessed how methylation at those sites affects ALDOB oligomerization and activity. Primary hepatocytes cultured with high/low glucose and/or fructose and wild type (WT) and Mat1a KO mice fed with a high-fructose diet examined AMPK-ALDOB signaling and MASLD progression. RESULTS In Mat1a KO livers ALDOB R173 is hypomethylated while ALDOB activity is enhanced. Recombinant ALDOB is methylated at R173 and R304 by protein arginine methyltransferase 1. Low hepatic SAMe level results in hypomethylated ALDOB, which favors the tetrameric form that has higher enzymatic activity, and higher capacity to signal to activate AMPK. Fructose, independently of glucose levels, inhibited AMPK activity and induced lipid accumulation in hepatocytes. Mat1a KO mice have hyperactivated AMPK and fructose feeding inhibits it, enhancing the accumulation of fat in the liver and the progression of MASLD. CONCLUSION Hepatic SAMe levels regulate ALDOB oligomeric state and enzymatic activity impacting on AMPK signaling and fructose-induced MASLD progression.
Collapse
Affiliation(s)
- Lucía Barbier-Torres
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - María Luque-Urbano
- CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), Technology Park of Bizkaia, 48160 Derio, Bizkaia, Spain
| | - Jyoti Chhimwal
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Aaron E Robinson
- Advanced Clinical Biosystems Research Institute, The Smidt Heart Institute, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA
| | - David Fernández-Ramos
- CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), Technology Park of Bizkaia, 48160 Derio, Bizkaia, Spain
| | - Fernando Lopitz-Otsoa
- CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), Technology Park of Bizkaia, 48160 Derio, Bizkaia, Spain
| | - Jennifer E Van Eyk
- Advanced Clinical Biosystems Research Institute, The Smidt Heart Institute, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Oscar Millet
- CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), Technology Park of Bizkaia, 48160 Derio, Bizkaia, Spain
| | - José M Mato
- CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), Technology Park of Bizkaia, 48160 Derio, Bizkaia, Spain.
| | - Shelly C Lu
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| |
Collapse
|
17
|
Chen M, Zhang S, Huang X, Zhang D, Zhu D, Ouyang C, Li Y. The protective effects and mechanism of myricetin in liver diseases (Review). Mol Med Rep 2025; 31:87. [PMID: 39917997 PMCID: PMC11811602 DOI: 10.3892/mmr.2025.13452] [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: 11/06/2024] [Accepted: 01/22/2025] [Indexed: 02/13/2025] Open
Abstract
Liver diseases have become one of the significant threats to global health. However, there is a lack of effective targeted therapeutic drugs in this field and the existing drugs used for liver disease treatment usually have side‑effects. Traditional Chinese medicine (TCM) has the distinctive advantages of multi‑target and low side‑effects. As a flavonoid with various pharmacological activities such as anti‑tumour, anti‑oxidant, anti‑inflammatory and anti‑bacterial, the TCM myricetin has been widely used in liver disease research. The present work focuses on the role and molecular mechanism of myricetin in liver diseases such as acute liver injury, fatty liver, liver fibrosis and hepatocellular carcinoma. It is a promising reference for further research and application of myricetin in the treatment of liver diseases.
Collapse
Affiliation(s)
- Mi Chen
- Hubei Key Laboratory of Diabetes and Angiopathy, School of Pharmacy, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Shengnan Zhang
- Hubei Key Laboratory of Diabetes and Angiopathy, School of Pharmacy, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
- School of Resources and Environmental Science and Engineering, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Xingqiong Huang
- Hubei Key Laboratory of Diabetes and Angiopathy, School of Pharmacy, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Dandan Zhang
- Hubei Key Laboratory of Diabetes and Angiopathy, School of Pharmacy, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Dan Zhu
- Hubei Key Laboratory of Diabetes and Angiopathy, School of Pharmacy, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Changhan Ouyang
- Hubei Key Laboratory of Diabetes and Angiopathy, School of Pharmacy, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Yankun Li
- Hubei Key Laboratory of Diabetes and Angiopathy, School of Pharmacy, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| |
Collapse
|
18
|
Wang X, Abu Bakar MH, Liqun S, Kassim MA, Shariff KA, Karunakaran T. Targeting metabolic diseases with celastrol: A comprehensive review of anti-inflammatory mechanisms and therapeutic potential. JOURNAL OF ETHNOPHARMACOLOGY 2025; 344:119560. [PMID: 40015541 DOI: 10.1016/j.jep.2025.119560] [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: 12/10/2024] [Revised: 02/15/2025] [Accepted: 02/23/2025] [Indexed: 03/01/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Tripterygium wilfordii is a traditional Chinese medicine used to treat rheumatic diseases, with properties such as clearing heat, detoxifying, dispelling wind, and relieving pain. In recent years, its active compound, celastrol, garnered significant attention for its potential therapeutic effects on metabolic diseases. Celastrol exhibits bioactivities such as regulating metabolic functions and anti-inflammatory effects, positioning it as a promising candidate for the treatment of obesity, diabetes, atherosclerosis (AS), and non-alcoholic fatty liver disease (NAFLD). AIM OF THE REVIEW This review aims to explore the pharmacological mechanisms of celastrol in metabolic diseases, focusing on its anti-inflammatory mechanisms and metabolic regulation effects, providing theoretical support for further investigation of its therapeutic potential in metabolic diseases. METHODS Literature was retrieved from PubMed, Web of Science, Scopus, Cochrane, and Google Scholar. This review primarily focuses on anti-inflammatory mechanisms of celastrol, its metabolic regulation, and toxicity studies, by systematically analyzing its effects in obesity, diabetes, AS, and NAFLD, providing scientific evidence for its potential clinical applications. RESULTS Celastrol regulates multiple signaling pathways, particularly inhibiting NF-κB and activating AMPK, reducing the production of pro-inflammatory cytokines and improving insulin sensitivity, enhancing its therapeutic potential in metabolic diseases. Additionally, celastrol regulates adipogenesis and energy metabolism by influencing key transcription factors such as PPARγ and SREBP-1c. Numerous studies highlight its role in alleviating oxidative stress and improving mitochondrial function, further enhancing its metabolic benefits. CONCLUSION In summary, celastrol holds great promise as a multi-target therapeutic agent for metabolic diseases, offering anti-inflammatory, metabolic regulatory, and antioxidative benefits. Despite these, challenges remain for the clinical application of celastrol due to its poor bioavailability and potential toxicity. Advanced formulation strategies and targeted delivery systems are urgently needed to overcome challenges related to bioavailability and clinical translation.
Collapse
Affiliation(s)
- Xiaojuan Wang
- Bioprocess Technology Division, School of Industrial Technology, Universiti Sains Malaysia, Gelugor, 11800, Penang, Malaysia; Department of Pharmacy, Taishan Vocational College of Nursing, 271099, Tai'an, Shandong, China
| | - Mohamad Hafizi Abu Bakar
- Bioprocess Technology Division, School of Industrial Technology, Universiti Sains Malaysia, Gelugor, 11800, Penang, Malaysia.
| | - Song Liqun
- Department of Pharmacy, Taishan Vocational College of Nursing, 271099, Tai'an, Shandong, China
| | - Mohd Asyraf Kassim
- Bioprocess Technology Division, School of Industrial Technology, Universiti Sains Malaysia, Gelugor, 11800, Penang, Malaysia
| | - Khairul Anuar Shariff
- School of Materials & Mineral Resources Engineering, Universiti Sains Malaysia, Nibong Tebal, 14300, Penang, Malaysia
| | | |
Collapse
|
19
|
Randeni N, Luo J, Xu B. Critical Review on Anti-Obesity Effects of Anthocyanins Through PI3K/Akt Signaling Pathways. Nutrients 2025; 17:1126. [PMID: 40218884 PMCID: PMC11990295 DOI: 10.3390/nu17071126] [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: 03/03/2025] [Revised: 03/18/2025] [Accepted: 03/23/2025] [Indexed: 04/14/2025] Open
Abstract
Obesity is a global health crisis and is one of the major reasons for the rising prevalence of metabolic disorders such as type 2 diabetes, cardiovascular diseases, and certain cancers. There has been growing interest in the search for natural molecules with potential anti-obesity effects; among the phytochemicals of interest are anthocyanins, which are flavonoid pigments present in many fruits and vegetables. Anthocyanins influence obesity via several signaling pathways. The PI3K/Akt signaling pathway plays a major role with a focus on downstream targets such as GLUT4, FOXO, GSK3β, and mTOR, which play a central role in the regulation of glucose metabolism, lipid storage, and adipogenesis. The influence of critical factors such as oxidative stress and inflammation also affect the pathophysiology of obesity. However, the studies reviewed have certain limitations, including variations in experimental models, bioavailability challenges, and a lack of extensive clinical validation. While anthocyanin shows tremendous potential, challenges such as poor bioavailability, stability, and regulatory matters must be overcome for successful functional food inclusion of anthocyanins. The future of anthocyanin-derived functional foods lies in their ability to overcome hurdles. Therefore, this review highlights the molecular mechanisms of obesity through the PI3K/Akt signaling pathways and explores how anthocyanins can modulate these signaling pathways to address obesity and related metabolic disorders. It also addresses some ways to solve the challenges, like bioavailability and stability, while emphasizing future possibilities for anthocyanin-based functional foods in obesity management.
Collapse
Affiliation(s)
| | | | - Baojun Xu
- Food Science and Technology Program, Department of Life Sciences, Beijing Normal-Hong Kong Baptist University, Zhuhai 519087, China; (N.R.); (J.L.)
| |
Collapse
|
20
|
Park YI, Park R, Lee S, Lee C, Yoo I, Ka H, Huh YH, Hong J, Park J. AMPK Knockout Impairs the Formation of Three-Dimensional Spheroids. Life (Basel) 2025; 15:525. [PMID: 40283080 PMCID: PMC12028351 DOI: 10.3390/life15040525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 03/19/2025] [Accepted: 03/20/2025] [Indexed: 04/29/2025] Open
Abstract
AMP-activated protein kinase (AMPK) is an important regulator of cellular energy homeostasis, and AMPK contributes to cell growth, apoptosis, and autophagy. Although most cell studies have been performed using two-dimensional (2D) cell culture, recent studies have demonstrated that the three-dimensional (3D) spheroid technique is helpful in various cell research fields, such as tumor biology, due to its resemblance to the 3D tissue structure. However, the role of AMPK in 3D spheroid formation has not been characterized clearly. This study used the AMPK knockout cell line to examine the role of AMPK in 3D spheroid formation and is the first report describing the generation of 3D spheroids using AMPK knockout cells. While control cells produced round spheroids with a similar length-to-width ratio, AMPK knockout produced an oval shape with a more significant length-to-width ratio. We demonstrate that AMPK knockout spheroids contain significantly more prominent lysosomes in each cell, indicating that autophagic flux is impaired in 3D spheroids. Finally, flow cytometry analysis showed that AMPK knockout spheroids contain more apoptotic cells than control cells. These results indicate that AMPK is required for efficient 3D spheroid formation.
Collapse
Affiliation(s)
- Yea-In Park
- Division of Biological Science and Technology, Yonsei University, Wonju 26493, Republic of Korea
| | - Rackhyun Park
- Department of Life Science, Yong-In University, Yongin 17092, Republic of Korea
| | - Siyun Lee
- Division of Biological Science and Technology, Yonsei University, Wonju 26493, Republic of Korea
| | - Chunghyeon Lee
- Division of Biological Science and Technology, Yonsei University, Wonju 26493, Republic of Korea
| | - Inkyu Yoo
- Division of Biological Science and Technology, Yonsei University, Wonju 26493, Republic of Korea
| | - Hakhyun Ka
- Division of Biological Science and Technology, Yonsei University, Wonju 26493, Republic of Korea
| | - Yang Hoon Huh
- Electron Microscopy Research Center, Korea Basic Science Institute (KBSI), Cheongju 28119, Republic of Korea
| | - Jongkwang Hong
- Division of Biological Science and Technology, Yonsei University, Wonju 26493, Republic of Korea
| | - Junsoo Park
- Division of Biological Science and Technology, Yonsei University, Wonju 26493, Republic of Korea
| |
Collapse
|
21
|
Zhang J, Wang Q, Zhou N, Liu J, Tao L, Peng Z, Hu G, Wang H, Fu L, Peng S. Fluorofenidone attenuates choline-deficient, l-amino acid-defined, high-fat diet-induced metabolic dysfunction-associated steatohepatitis in mice. Sci Rep 2025; 15:9863. [PMID: 40118958 PMCID: PMC11928590 DOI: 10.1038/s41598-025-94401-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 03/13/2025] [Indexed: 03/24/2025] Open
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH), a severe form of metabolic dysfunction-associated steatotic liver disease (MASLD), involves hepatic lipid accumulation, inflammation, and fibrosis. It can progress to cirrhosis or hepatocellular carcinoma without timely treatment. Current treatment options for MASH are limited. This study explores the therapeutic effects of fluorofenidone (AKF-PD), a novel small-molecule compound with antifibrotic and anti-inflammatory properties, on MASH in mouse model. Mice fed a choline-deficient, l-amino acid-defined, high-fat diet (CDAHFD) were treated with AKF-PD, resulting in reduced serum ALT, AST, hepatic lipid accumulation, liver inflammation, and fibrosis. Network pharmacology and RNA-sequencing analyses suggested that AKF-PD influenced multiple metabolic, inflammatory, and fibrosis-related pathways. Further experiments verified that AKF-PD activated hepatic AMPK signaling, leading to the inhibition of the downstream SREBF1/SCD1 pathway and the activation of autophagy. Additionally, AKF-PD suppressed the expression of various inflammatory factors, reduced macrophage infiltration, and inhibited NLRP3 inflammasome activation. Moreover, AKF-PD attenuated liver fibrosis by inhibiting TGFβ1/SMAD signaling. In conclusion, this study reveals that AKF-PD effectively decreases hepatic lipid accumulation, liver inflammation and fibrosis in a CDAHFD-induced MASH model, positioning AKF-PD as a promising candidate for the treatment of MASH.
Collapse
Affiliation(s)
- Jian Zhang
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Qianbing Wang
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Nianqi Zhou
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Jinqing Liu
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Lijian Tao
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Zhangzhe Peng
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Gaoyun Hu
- Faculty of Pharmaceutical Sciences, Central South University, Changsha, 410008, Hunan, China
| | - Huiwen Wang
- Department of Infection Control Center, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| | - Lei Fu
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| | - Shifang Peng
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
22
|
Yu J, Naseem S, Park S, Hur S, Choi Y, Lee T, Li X, Choi S. FASN, SCD, and PLAG1 Gene Polymorphism and Association with Carcass Traits and Fatty Acid Profile in Hanwoo Cattle. Animals (Basel) 2025; 15:897. [PMID: 40150426 PMCID: PMC11939486 DOI: 10.3390/ani15060897] [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: 01/27/2025] [Revised: 03/18/2025] [Accepted: 03/19/2025] [Indexed: 03/29/2025] Open
Abstract
Genetic polymorphisms have a great impact on enhancing quantitative traits in cattle. In this study, Fatty acid synthase (FASN) g. 16024 (A>G), Stearoyl-CoA desaturase (SCD) g. 10329 (C>T), and pleomorphic adenoma gene (PLAG1) g. 25003338 (C>G) genotypic and allelic polymorphisms were evaluated, along with their associations with fatty acid composition, adipogenic gene expression, and carcass characteristics (carcass weight, yield grade, backfat thickness, and marbling score) in Hanwoo steers. A total of 128 Hanwoo steers were selected for this study and the Polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) method was used to identify polymorphism of these genes. The AG genotype and G allele in FASN g. 16024 (A>G), CT genotype and T allele in SCD g. 10329 (C>T), and GG genotype and G allele in PLAG1 g. 25003338 (C>G) showed higher frequency and positively correlated with carcass traits, yield, and quality grades. Fatty acid composition results indicate that C18:3n-6, C20:1, and C20:2n-6 were significantly higher in the AA genotype of FASN gene, C14:1 and C18:3n-6 in the CC genotype, and C16:1 in the TT genotype of SCD gene. C12:0, C14:0, C16:1, C18:0, and C20:0 were higher in the CC genotype of PLAG1 gene. Furthermore, RT-qPCR analysis of adipogenesis-related genes (AMP-activated protein kinase-α (AMPKα), Carnitine palmitoyl transferase-1β (CPT1), G-coupled protein receptor-43 (GPR43), and SCD) across different SNP genotypes suggests a systemic interaction between genetic factors and adipogenesis in beef cattle. This study emphasizes the significance of FASN g. 16024 (A>G), SCD g. 10329 (C>T), and PLAG1 g. 25003338 (C>G) SNPs for genetic selection to enhance beef quality and elucidate lipid metabolic pathways in Hanwoo cattle.
Collapse
Affiliation(s)
- Jia Yu
- Department of Animal Science, Chungbuk National University, Cheongju 28644, Republic of Korea; (J.Y.); (Y.C.); (T.L.)
| | - Sajida Naseem
- Engineering Research Center of North-East Cold Region Beef Cattle Science & Technology Innovation Ministry of Education, Department of Animal Science, Yanbian University, Yanji 133002, China;
| | - Sungkwon Park
- Department of Food Science and Biotechnology, Sejong University, Seoul 05006, Republic of Korea;
| | - Sunjin Hur
- Department of Animal Science and Technology, Chung-Ang University, Anseong 17546, Republic of Korea;
| | - Yoonbin Choi
- Department of Animal Science, Chungbuk National University, Cheongju 28644, Republic of Korea; (J.Y.); (Y.C.); (T.L.)
| | - Teahyung Lee
- Department of Animal Science, Chungbuk National University, Cheongju 28644, Republic of Korea; (J.Y.); (Y.C.); (T.L.)
| | - Xiangzi Li
- Engineering Research Center of North-East Cold Region Beef Cattle Science & Technology Innovation Ministry of Education, Department of Animal Science, Yanbian University, Yanji 133002, China;
| | - Seongho Choi
- Department of Animal Science, Chungbuk National University, Cheongju 28644, Republic of Korea; (J.Y.); (Y.C.); (T.L.)
| |
Collapse
|
23
|
Berdowska I, Matusiewicz M, Fecka I. A Comprehensive Review of Metabolic Dysfunction-Associated Steatotic Liver Disease: Its Mechanistic Development Focusing on Methylglyoxal and Counterbalancing Treatment Strategies. Int J Mol Sci 2025; 26:2394. [PMID: 40141037 PMCID: PMC11942149 DOI: 10.3390/ijms26062394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/21/2025] [Accepted: 03/04/2025] [Indexed: 03/28/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a multifactorial disorder characterized by excessive lipid accumulation in the liver which dysregulates the organ's function. The key contributor to MASLD development is insulin resistance (IR) which affects many organs (including adipose tissue, skeletal muscles, and the liver), whereas the molecular background is associated with oxidative, nitrosative, and carbonyl stress. Among molecules responsible for carbonyl stress effects, methylglyoxal (MGO) seems to play a major pathological function. MGO-a by-product of glycolysis, fructolysis, and lipolysis (from glycerol and fatty acids-derived ketone bodies)-is implicated in hyperglycemia, hyperlipidemia, obesity, type 2 diabetes, hypertension, and cardiovascular diseases. Its causative effect in the stimulation of prooxidative and proinflammatory pathways has been well documented. Since metabolic dysregulation leading to these pathologies promotes MASLD, the role of MGO in MASLD is addressed in this review. Potential MGO participation in the mechanism of MASLD development is discussed in regard to its role in different signaling routes leading to pathological events accelerating the disorder. Moreover, treatment strategies including approved and potential therapies in MASLD are overviewed and discussed in this review. Among them, medications aimed at attenuating MGO-induced pathological processes are addressed.
Collapse
Affiliation(s)
- Izabela Berdowska
- Department of Medical Biochemistry, Faculty of Medicine, Wroclaw Medical University, Chałubińskiego 10, 50-368 Wrocław, Poland;
| | - Małgorzata Matusiewicz
- Department of Medical Biochemistry, Faculty of Medicine, Wroclaw Medical University, Chałubińskiego 10, 50-368 Wrocław, Poland;
| | - Izabela Fecka
- Department of Pharmacognosy and Herbal Medicines, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wrocław, Poland
| |
Collapse
|
24
|
Karimzadeh K, Unniappan S, Zahmatkesh A. Spirulina platensis Peptide-Loaded Nanoliposomes Alleviate Hepatic Lipid Accumulation in Male Wistar Rats by Influencing Redox Homeostasis and Lipid Metabolism via the AMPK Signaling Pathway. Appl Biochem Biotechnol 2025; 197:1696-1725. [PMID: 39601973 DOI: 10.1007/s12010-024-05089-w] [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] [Accepted: 11/12/2024] [Indexed: 11/29/2024]
Abstract
Spirulina platensis low-molecular-weight peptides (SP) have been reported to exhibit antioxidant and hepatoprotective properties. However, the limited bioavailability and solubility of SPs limit their potential applications. In this study, to examine the potential anti-obesity effects and underlying mechanisms of SPs, high-fat diet-induced non-alcoholic fatty liver disease (NAFLD) model rats were treated with SPs and SP-loaded nanoliposomes. Furthermore, hepatic biochemical parameters, inflammatory markers, histopathological changes, and genes involved in AMPK signaling were analyzed. SP-loaded nanoliposomes demonstrated a spherical shape with slower and sustained SP release. SP and SP-loaded nanoliposomes mitigated hepatic damage by lowering serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) and increasing hepatic antioxidant enzymes, which are manifested in improving histopathological findings. In addition, notably, SP-loaded nanoliposomes downregulated lipogenic fatty acid synthase (FAS) and sterol regulatory element-binding protein-1c (SREBP-1c) in the liver. Meanwhile, an upregulation of phosphorylated AMP-activated protein kinase (P-AMPK), lipid acid oxidation-related genes carnitine palmitoyltransferase-1 (CPT-1), and peroxisome proliferator-activated receptor alpha (PPAR-α) was found in the rat liver. This data implies that SP and SP-loaded nanoliposomes exhibit protective potential in rats against the HFD-induced NAFLD, which is mediated through the activation of the AMPK signaling pathway.
Collapse
Affiliation(s)
- Katayoon Karimzadeh
- Laboratory of Integrative Neuroendocrinology, Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Canada.
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Canada.
| | - Suraj Unniappan
- Laboratory of Integrative Neuroendocrinology, Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Asgar Zahmatkesh
- Aquaculture Department, Gilan Agricultural and Natural Resources Research and Education Center, AREEO, Rasht, Iran
| |
Collapse
|
25
|
Nawaz H, Lee H, Kang S, Kim H, Kim W, Go GW. Alternate-day fasting enhanced weight loss and metabolic benefits over pair-fed calorie restriction in obese mice. Obesity (Silver Spring) 2025; 33:512-521. [PMID: 39905657 DOI: 10.1002/oby.24211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 10/25/2024] [Accepted: 11/01/2024] [Indexed: 02/06/2025]
Abstract
OBJECTIVE Both alternate-day fasting (ADF) and calorie restriction (CR) are effective weight loss strategies. However, most individuals find it difficult to adhere to CR. Furthermore, CR can induce an excessive loss of not only fat but also muscle mass. This study aimed to compare the effects of ADF and pair-feeding (PF) CR on metabolic pathways underlying obesity in mice with high-fat diet (HFD)-induced obesity. METHODS Male C57BL/6N Tac mice (n = 10 per group) were fed an HFD for 8 weeks to establish a diet-induced obesity model. Mice were then continued on the HFD with either alternate-day access to food or PF for the next 8 weeks. We measured body weight, adiposity, plasma biomarkers, and molecular mechanisms involving lipolysis and autophagy. RESULTS Both ADF and PF resulted in comparable weight and fat loss. Compared with PF, ADF showed a significant reduction in liver weight and hepatic triglyceride levels. ADF significantly increased plasma ketone body levels and white adipose tissue lipolysis. Compared with PF, ADF tended to activate autophagy elongation and autophagosome formation, which were insignificant. CONCLUSIONS These findings indicated that ADF is a promising intervention for metabolic diseases, potentially due to its superior efficacy in promoting ketogenesis and lipolysis compared with PF.
Collapse
Affiliation(s)
- Hadia Nawaz
- Department of Food and Nutrition, Hanyang University, Seoul, Republic of Korea
| | - Haneul Lee
- Department of Food and Nutrition, Hanyang University, Seoul, Republic of Korea
| | - Sumin Kang
- Department of Food and Nutrition, Hanyang University, Seoul, Republic of Korea
| | - Hayoon Kim
- Department of Food and Nutrition, Hanyang University, Seoul, Republic of Korea
| | - Wooki Kim
- Department of Food and Nutrition, Yonsei University, Seoul, Republic of Korea
| | - Gwang-Woong Go
- Department of Food and Nutrition, Hanyang University, Seoul, Republic of Korea
| |
Collapse
|
26
|
Deng X, Liu Z, Yang S. β-Elemene inhibits adipogenesis in 3T3-L1 cells by regulating AMPK pathway. J Clin Biochem Nutr 2025; 76:125-130. [PMID: 40151406 PMCID: PMC11936742 DOI: 10.3164/jcbn.24-179] [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: 10/16/2024] [Accepted: 11/16/2024] [Indexed: 03/29/2025] Open
Abstract
The prevalence of childhood obesity in global is quickly augmented, resulting into grievous public health problems and influencing adolescent development. β-Elemene is a sesquiterpene, and can extracted from traditional Chinese medicine-Curcuma longa L. β-Elemene has been discovered to display regulatory functions in multiple diseases, but it's roles in obesity need further investigations. The purpose of this work is to investigate the regulatory impacts of β-elemene on obesity progression and associated pathways. In this study, it was revealed that the heightened lipid accumulation in 3T3-L1 cells triggered by 3-isobutyl-1-methylxanthine + dexamethazone + insulin (MDI) can be restrained by β-elemene. Furthermore, β-elemene can modulate lipid metabolism in 3T3-L1 cells mediated by MDI. The glucose consumption was descended after insulin resistance treatment, but this impact was reversed after β-elemene treatment. At last, it was illustrated that the AMPK pathway was retarded after β-elemene induction, but this change was offset after β-elemene treatment. To sum up, our results manifested that β-elemene inhibited adipogenesis in 3T3-L1 cells, and evoked the AMPK pathway. This project may supply serviceable insights of β-elemene in the progression of obesity.
Collapse
Affiliation(s)
- Xiang Deng
- Department of Pediatrics, Chengdu Fifth People’s Hospital, No. 33, Mashi street, Wenjiang District, Chengdu, Sichuan 611130, China
| | - Zhenmin Liu
- Department of Pediatrics, Chengdu Fifth People’s Hospital, No. 33, Mashi street, Wenjiang District, Chengdu, Sichuan 611130, China
| | - Sen Yang
- Department of Pediatrics, Chengdu Fifth People’s Hospital, No. 33, Mashi street, Wenjiang District, Chengdu, Sichuan 611130, China
- Department of Pediatrics, School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, 611137, China
| |
Collapse
|
27
|
Song L, Huang Y, Liu L, Chang X, Hu L, Wang G, Xu L, Zhang T, Wang Y, Xiao Y, Yang H, Ran S, Shi Q, Wang T, Shi M, Zhou Y, Guo B. Meteorin-like alleviates hepatic steatosis by regulating hepatic triglyceride secretion and fatty acid oxidation. Cell Rep 2025; 44:115246. [PMID: 39918960 DOI: 10.1016/j.celrep.2025.115246] [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: 10/02/2024] [Revised: 11/29/2024] [Accepted: 01/09/2025] [Indexed: 02/09/2025] Open
Abstract
Amid a rising prevalence of non-alcoholic fatty liver disease (NAFLD), there is still an unmet need to better treat it. We identified a secreted factor, Meteorin-like (Metrnl), with decreased levels in livers with hepatic steatosis. Notably, recombinant Metrnl ameliorated hepatic steatosis in NAFLD mouse models. Mechanistically, Metrnl exerted dual effects by promoting triglyceride (TG) transportation by the phosphatidylinositol 3-kinase (PI3K)/Akt/Sp1/cytidylyltransferase α (CCTα) axis, thereby increasing the biosynthesis of phosphatidylcholine (PC) to facilitate TG secretion from the liver while facilitating AMP-activated protein kinase (AMPK)-dependent fatty acid oxidation (FAO). Exogenous injection of cytidine diphosphocholine (CDP)-choline, the production of CCTα, to increase PC synthesis, was shown to restore the inhibition of TG secretion in hepatic Metrnl-deficient (LKO-Met) mice. Combining CDP-choline and an AMPK activator was sufficient to rescue hepatic steatosis in LKO-Met mice. Collectively, these findings reveal unexpected roles of Metrnl as a factor in PC biosynthesis, TG secretion, and FAO, suggesting potential therapeutic application for NAFLD.
Collapse
Affiliation(s)
- Lingyu Song
- Department of Pathophysiology, and Department of Gastroenterology of the Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Gui'an New Area, 561113, China
| | - Yali Huang
- Department of Pathophysiology, and Department of Gastroenterology of the Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Gui'an New Area, 561113, China
| | - Lu Liu
- Department of Pathophysiology, and Department of Gastroenterology of the Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Gui'an New Area, 561113, China
| | - Xuebing Chang
- Department of Pathophysiology, and Department of Gastroenterology of the Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Gui'an New Area, 561113, China
| | - Laying Hu
- Department of Pathophysiology, and Department of Gastroenterology of the Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Gui'an New Area, 561113, China
| | - Guifang Wang
- Department of Pathophysiology, and Department of Gastroenterology of the Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Gui'an New Area, 561113, China
| | - Lifen Xu
- Department of Pathology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou, China
| | - Tian Zhang
- Department of Pathophysiology, and Department of Gastroenterology of the Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Gui'an New Area, 561113, China
| | - Yuanyuan Wang
- Department of Pathophysiology, and Department of Gastroenterology of the Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Gui'an New Area, 561113, China
| | - Ying Xiao
- Department of Pathophysiology, and Department of Gastroenterology of the Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Gui'an New Area, 561113, China
| | - Hong Yang
- Department of Pathophysiology, and Department of Gastroenterology of the Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Gui'an New Area, 561113, China
| | - Suye Ran
- Department of Pathophysiology, and Department of Gastroenterology of the Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Gui'an New Area, 561113, China
| | - Qing Shi
- Department of Pathophysiology, and Department of Gastroenterology of the Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Gui'an New Area, 561113, China
| | - Tuanlao Wang
- School of Pharmaceutical Sciences, State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, 361005, Fujian, China
| | - Mingjun Shi
- Department of Pathophysiology, and Department of Gastroenterology of the Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Gui'an New Area, 561113, China.
| | - Yuxia Zhou
- Department of Pathophysiology, and Department of Gastroenterology of the Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Gui'an New Area, 561113, China.
| | - Bing Guo
- Department of Pathophysiology, and Department of Gastroenterology of the Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Gui'an New Area, 561113, China; Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases and Guizhou Province Talent Base of Research on the Pathogenesis and Drug Prevention and Treatment for Common Major Diseases, Guizhou Medical University, Gui'an New Area, 561113, China; Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, Guizhou Medical University, Gui'an New Area, 561113, Guizhou, China.
| |
Collapse
|
28
|
Lin YL, Sutopo CCY, Kao YF, Tseng JK, Chen YC. Modulatory Effects of Ophiocordyceps sinensis Mycelia on Hepatosteatosis Development in a High-Fat Dietary Habit. ENVIRONMENTAL TOXICOLOGY 2025. [PMID: 39980443 DOI: 10.1002/tox.24499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 01/21/2025] [Accepted: 02/10/2025] [Indexed: 02/22/2025]
Abstract
The global rise in obesity is closely associated with the increasing prevalence of nonalcoholic fatty liver disease (NAFLD) and metabolic syndromes, posing significant health challenges. This study explored the ameliorative effects of Ophiocordyceps sinensis mycelia (TCM-NA01 formula: 1.4 mg adenosine and 55.2 mg polysaccharide/capsule) on hepatosteatosis development in a high-fat diet (HFD)-fed mice. TCM-NA01 supplementation significantly reduced (p < 0.05) body weight, adipose tissue, serum triglyceride (TG)/cholesterol (TC), aspartate aminotransferase (AST), and alanine aminotransferase (ALT), liver TG/TC levels in HFD-fed mice. Increased (p < 0.05) fecal-lipid and bile-acid outputs were observed. Apparent reductions in lipid-droplet and steatosis scores (p < 0.05) in the HFD-fed mice supplemented with TCM-NA01. Furthermore, TCM-NA01 modulated lipid metabolism by decreasing fatty acid synthesis and promoting fatty acid β-oxidation. TCM-NA01 also enhanced liver antioxidant capacity and decreased proinflammatory cytokines (p < 0.05). These findings underscore the potential of O. sinensis mycelia as a nutraceutical agent for alleviating hepatosteatosis, liver oxidative stress, and chronic inflammation, offering a promising strategy for the management of obesity and NAFLD.
Collapse
Affiliation(s)
- Yi-Ling Lin
- Department of Animal Science and Technology, National Taiwan University, Taipei City, Taiwan
- Undergraduate and Graduate Programs of Nutrition Science, National Taiwan Normal University, Taipei City, Taiwan
| | | | - Yi-Feng Kao
- Seafood Technology Division, Fisheries Research Institute, Ministry of Agriculture, Keelung City, Taiwan
| | - Jung-Kai Tseng
- Department of Optometry, Asia University, Taichung, Taiwan
| | - Yi-Chen Chen
- Department of Animal Science and Technology, National Taiwan University, Taipei City, Taiwan
- Master Program in Global Agriculture Technology and Genomic Science, International College, National Taiwan University, Taipei City, Taiwan
| |
Collapse
|
29
|
Guo Y, Guo W, Chen H, Sun J, Yin Y. Mechanisms of sepsis-induced acute liver injury: a comprehensive review. Front Cell Infect Microbiol 2025; 15:1504223. [PMID: 40061452 PMCID: PMC11885285 DOI: 10.3389/fcimb.2025.1504223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 01/31/2025] [Indexed: 05/13/2025] Open
Abstract
Sepsis is a severe, often life-threatening form of organ dysfunction that arises from an inappropriately regulated host response to infectious pathogen exposure. As the largest gland in the body, the liver serves as a regulatory hub for metabolic, immune, and detoxification activity. It is also an early sepsis target organ such that hepatic dysfunction is observed in 34-46% of patients with sepsis. The precise mechanisms that give rise to sepsis-induced liver injury, however, remain incompletely understood. Based on the research conducted to date, dysregulated systemic inflammation, microbial translocation, microcirculatory abnormalities, cell death, metabolic dysfunction, and liver inflammation may all contribute to the liver damage that can arise in the context of septicemia. This review was developed to provide an overview summarizing the potential mechanisms underlying sepsis-induced liver injury, informing the selection of potential targets for therapeutic intervention and providing a framework for the alleviation of patient symptoms and the improvement of prognostic outcomes.
Collapse
Affiliation(s)
- Yongjing Guo
- Department of Emergency and Critical Care, the Second Hospital of Jilin University, Changchun, China
| | - Wanxu Guo
- Department of Neonate, The Second Hospital of Jilin University, Changchun, China
| | - Huimin Chen
- Department of Neonate, The Second Hospital of Jilin University, Changchun, China
| | - Jian Sun
- Department of Emergency and Critical Care, the Second Hospital of Jilin University, Changchun, China
| | - Yongjie Yin
- Department of Emergency and Critical Care, the Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
30
|
Lee HL, Kim JM, Go MJ, Lee HS, Kim JH, Kim IY, Seong GS, Heo HJ. Fermented Protaetia brevitarsis Larvae Alleviates High-Fat Diet-Induced Non-Alcoholic Fatty Liver Disease in C57BL/6 Mice via Regulation of Lipid Accumulation and Inflammation. J Microbiol Biotechnol 2025; 35:e2409025. [PMID: 39947694 PMCID: PMC11876019 DOI: 10.4014/jmb.2409.09025] [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: 09/12/2024] [Revised: 12/11/2024] [Accepted: 12/17/2024] [Indexed: 03/06/2025]
Abstract
Non-alcoholic fatty liver disease (NAFLD), characterized by hepatic steatosis and hepatitis, is the most frequently encountered complication of type 2 diabetes mellitus (T2DM). Due to its hepatoprotective, anti-obesity, antioxidant, and anti-inflammatory effects, Protaetia brevitarsis (P. brevitarsis) larvae have been used as traditional medicine to treat liver diseases since ancient times. Therefore, this study was conducted to confirm the positive effect of fermented P. brevitarsis larvae (FPB) on NAFLD. The results showed that high-fat diet (HFD)-induced dysglycemia was improved by treatment with FPB as determined by testing for fasting blood glucose and oral glucose tolerance. The weight of liver and white adipose tissue and the levels of serum lipid, hepatotoxicity, and nephrotoxicity indicators were reduced by FPB. In addition, oxidative stress and mitochondrial dysfunction caused by HFD were improved by FPB. In a similar manner, HFD-induced hepatic steatosis was prevented by FPB through regulation of the AMP-activated protein kinase pathway and serum lipid profile. HFD-induced hepatitis and apoptosis were ameliorated by FPB via the nuclear factor-kappa B pathway and the B-cell lymphoma 2 protein family. In conclusion, this study suggests the potential for application of FPB as a prophylactic agent for treatment of NAFLD through suppression of lipid accumulation and inflammation in the liver.
Collapse
Affiliation(s)
- Hyo Lim Lee
- Division of Applied Life Science (BK21), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Jong Min Kim
- Division of Applied Life Science (BK21), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Min Ji Go
- Division of Applied Life Science (BK21), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Han Su Lee
- Division of Applied Life Science (BK21), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Ju Hui Kim
- Division of Applied Life Science (BK21), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - In Young Kim
- Division of Applied Life Science (BK21), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Geum-Su Seong
- Korea Food Research Institute (KFRI), Wanju Zipcode, Republic of Korea
| | - Ho Jin Heo
- Division of Applied Life Science (BK21), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea
| |
Collapse
|
31
|
Ezhilarasan D, Karthick M, Sharmila M, Sanjay S, Mani U. Deciphering the Molecular Mechanisms of HAART-Induced Hepatotoxicity. J Biochem Mol Toxicol 2025; 39:e70174. [PMID: 39959953 DOI: 10.1002/jbt.70174] [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: 07/23/2024] [Revised: 11/19/2024] [Accepted: 02/06/2025] [Indexed: 05/09/2025]
Abstract
Highly active antiretroviral therapy (HAART), consisting of three or more antiretroviral drugs, is recommended for patients with HIV infection. HAART effectively reduces HIV RNA levels, lowers the risk of opportunistic infections, and improves immune function and survival rates. However, it is also associated with an increased risk of liver injury in HIV-infected individuals. This review aims to summarize the mechanisms underlying HAART-induced liver injury. A comprehensive search was conducted in PubMed and EMBASE using keywords such as "Antiretroviral/ARV drugs and drug-induced liver injury (DILI)," "HAART and DILI," "Antiretroviral therapy and DILI," and "HIV infection and DILI." Relevant papers published before March 2024 were included. Experimental studies have demonstrated that zidovudine and efavirenz can cause structural alterations in mitochondria, impair the respiratory chain, generate free radicals, and deplete mitochondrial DNA, leading to oxidative and endoplasmic reticulum stress, as well as the accumulation of advanced glycation end products in liver tissue. Zidovudine disrupts lipid homeostasis by increasing fatty acid synthesis and reducing metabolism. Efavirenz and its metabolite, 8-hydroxyefavirenz, induce hepatocellular death and activate proapoptotic markers through c-Jun N-terminal kinase signaling. Additionally, lamivudine has been shown to induce liver injury and oxidative stress in rats. Clinically, approximately 50% of HIV patients on HAART regimens containing non-nucleoside reverse transcriptase inhibitors experience mild to moderate liver injury. HAART regimens that include efavirenz, lamivudine, and tenofovir impair glucose and lipid homeostasis in rats, highlighting the need for caution in HIV patients with fatty liver disease. Patients with viral hepatitis coinfection, those taking antitubercular drugs or cotrimoxazole, and those on nevirapine-containing regimens are at particularly high risk. Regular monitoring of liver function is essential to prevent liver damage associated with HAART in HIV-infected patients. While HAART significantly improves survival rates in HIV patients, it also poses a considerable risk of liver injury, necessitating careful monitoring and management.
Collapse
Affiliation(s)
- Devaraj Ezhilarasan
- Department of Pharmacology, Hepatology and Molecular Medicine Lab, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, India
| | - Munusamy Karthick
- Department of Pharmacology, Hepatology and Molecular Medicine Lab, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, India
| | - Muthusethupathi Sharmila
- Department of Pharmacology, Hepatology and Molecular Medicine Lab, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, India
| | - Somasundaram Sanjay
- Department of Pharmacology, Hepatology and Molecular Medicine Lab, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, India
| | - Uthirappan Mani
- Animal House Division, CSIR-Central Leather Research Institute, Adyar, Chennai, India
| |
Collapse
|
32
|
Wu M, Li K, Wu J, Ding X, Ma X, Wang W, Xiao W. Ginsenoside Rg1: A bioactive therapeutic agent for diverse liver diseases. Pharmacol Res 2025; 212:107571. [PMID: 39756553 DOI: 10.1016/j.phrs.2024.107571] [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: 09/07/2024] [Revised: 12/10/2024] [Accepted: 12/29/2024] [Indexed: 01/07/2025]
Abstract
Diverse liver diseases are characterised by late diagnosis and rapid progression and have become one of the major threats to human health. To delay the transition from benign tissue lesions to a substantial organ injury, scientists have gradually applied natural compounds derived from plants as a complementary therapy in the field of hepatology. Ginseng (Panax ginseng C. A. Meyer) is a tonic traditional Chinese herbal medicine, and natural products, including ginsenoside Rg1 (G-Rg1), which is a kind of 20(S)-protopanaxatriol saponin with a relatively high biological activity, can be isolated from the roots or stems of ginseng. Given these information, this review aimed to summarise and discuss the metabolic mechanisms of G-Rg1 in the regulation of diverse liver diseases and the measures to improve its bioavailability. As a kind of monomer in Chinese medicine with multitarget pharmacological effects, G-Rg1 can provide significant therapeutic benefits in the alleviation of alcoholic liver disease, nonalcoholic fatty liver disease, liver fibrosis, viral hepatitis, etc., which mainly rely on the inhibition of apoptosis, strengthening endogenous anti-inflammatory and antioxidant mechanisms, activation of immune responses and regulation of efflux transport signals, to improve pathological changes in the liver caused by lipid deposition, inflammation, oxidative stress, accumulation of hepatotoxic product, etc. However, the poor bioavailability of G-Rg1 must be overcome to improve its clinical application value. In summary, focusing on the hepatoprotective benefits of G-Rg1 will provide new insights into the development of natural Chinese medicine resources and their pharmaceutical products to target the treatment of liver diseases.
Collapse
Affiliation(s)
- Mingyu Wu
- Shanghai Key Lab of Human Performance (Shanghai University of sport), Shanghai University of Sport, Shanghai 200438, China; The Key Lab of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China.
| | - Ke Li
- Shanghai Key Lab of Human Performance (Shanghai University of sport), Shanghai University of Sport, Shanghai 200438, China; The Key Lab of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China.
| | - Jiabin Wu
- Shanghai Key Lab of Human Performance (Shanghai University of sport), Shanghai University of Sport, Shanghai 200438, China; The Key Lab of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China.
| | - Xianyi Ding
- Shanghai Key Lab of Human Performance (Shanghai University of sport), Shanghai University of Sport, Shanghai 200438, China; The Key Lab of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China.
| | - Xiaotong Ma
- Shanghai Key Lab of Human Performance (Shanghai University of sport), Shanghai University of Sport, Shanghai 200438, China; The Key Lab of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China.
| | - Wenhong Wang
- Shanghai Key Lab of Human Performance (Shanghai University of sport), Shanghai University of Sport, Shanghai 200438, China; Biomedical Research Institute, Hunan University of Medicine, Huaihua 418000, China; The Key Lab of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China.
| | - Weihua Xiao
- Shanghai Key Lab of Human Performance (Shanghai University of sport), Shanghai University of Sport, Shanghai 200438, China; The Key Lab of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China.
| |
Collapse
|
33
|
Zhang L, Zheng Y, Shao M, Chen A, Liu M, Sun W, Li T, Fang Y, Dong Y, Zhao S, Luo H, Feng J, Wang Q, Li L, Zheng Y. AlphaFold-based AI docking reveals AMPK/SIRT1-TFEB pathway modulation by traditional Chinese medicine in metabolic-associated fatty liver disease. Pharmacol Res 2025; 212:107617. [PMID: 39832686 DOI: 10.1016/j.phrs.2025.107617] [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: 12/04/2024] [Revised: 01/12/2025] [Accepted: 01/17/2025] [Indexed: 01/22/2025]
Abstract
Metabolic-associated fatty liver disease (MAFLD) is a chronic, progressive disorder characterized by hepatic steatosis and excessive lipid accumulation. Its high global adult prevalence (approximately 50.7 %) is a significant concern worldwide. However, FDA-approved therapeutic drugs remains lacking. Qigui Jiangzhi Formula (QGJZF) shows promise in treating MAFLD by effectively decreasing lipid levels and improving hepatic steatosis, however its mechanisms remain unclear. This study investigated QGJZF's effects in high-fat diet-induced zebrafish and golden hamsters, and in palmitate (PA) and oleic acid (OA) - induced HepG2 cells, using the SymMap database to identify potential targets and pathways of QGJZF in MAFLD and AlphaFold algorithms to predict protein structures. In vivo, QGJZF significantly alleviated hepatic lipid deposition. Intriguingly, QGJZF decreased lipid droplets and its levels are negative correlated with the numbers of autolysosomes, indicating that QGJZF's mechanism of ameliorating liver lipid deposition may be related to the regulation of autophagy. QGJZF upregulated the expressions of phosphorylated -Adenosine 5'-monophosphate (AMP) - activated protein kinase (p-AMPK), Sirtuin deacetylase 1 (SIRT1) and Transcription factor EB (TFEB), accompanied by the changes in autophagy-related proteins. In vitro, QGJZF inhibited the lipid deposition in PA/OA-stimulated HepG2 cells, and its effect was blocked by an autophagy inhibitor Baf-A1, which was mediated through upregulation of TFEB and its mediated autophagy-lysosomal pathway. Moreover, cotreatment with AMPK inhibitor Compound C, the regulation of QGJZF on TFEB, SIRT1, autophagy-related protein levels, and lipid deposition were reversed. Network pharmacology identified the PRKAA2 (AMPK) and SIRT1 as key hub targets. Futher analysis of their structures using AlphaFold3 algorithms, yielded high-ranking scores of 0.97 and 0.93, respectively. Liquid chromatography-mass spectrometry combined with molecular docking expounded its five compounds in QGJZF binding to AMPK protein. These findings suggest that QGJZF as a therapeutic agent in augmenting autophagy-facilitated lipid clearance for the management of MAFLD via AMPK/SIRT1-TFEB axis.
Collapse
Affiliation(s)
- Lulu Zhang
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing 100029, China; Shanxi University of Chinese Medicine, Jinzhong, Shanxi 030619, China
| | - Yi Zheng
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing 100029, China; School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Mingyan Shao
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Aiping Chen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Meiyi Liu
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing 100029, China; School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Wenlong Sun
- Institute of Biomedical Research, School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255000, China
| | - Tianxing Li
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yini Fang
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yang Dong
- Monitoning and Statistical Research Center, National Administration of Traditional Chinese Medicine, Beijing 100600, China
| | - Shipeng Zhao
- Graduate School of China Academy of Chinese Medical Sciences, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Hui Luo
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Juan Feng
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen 518118, China.
| | - Qi Wang
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Lingru Li
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Yanfei Zheng
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing 100029, China.
| |
Collapse
|
34
|
Katoch S, Chhimwal J, Singh D, Kumar D, Patial V. Picrosides-rich fraction from Picrorhiza kurroa attenuates steatohepatitis in zebrafish and mice by modulating lipid metabolism and inflammation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 137:156368. [PMID: 39827774 DOI: 10.1016/j.phymed.2025.156368] [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: 09/25/2023] [Revised: 12/23/2024] [Accepted: 01/01/2025] [Indexed: 01/22/2025]
Abstract
BACKGROUND Non-alcoholic steatohepatitis (NASH) has become a serious public health concern with high global prevalence. The lack of safe and efficient treatment for the condition demands exploring new therapeutic solutions. PURPOSE In the present study, we investigated the protective efficacy of picrosides-rich fraction (PF) from Picrorhiza kurroa against steatohepatitis and revealed the molecular mechanism of action. METHODS PF was prepared and characterized using UPLC analysis. Initially, the efficacy of PF was studied on the zebrafish model of NASH. Further, a Methionine and Choline-Deficient (MCD) diet-induced NASH model in mice was employed to evaluate the hepatoprotective efficacy of PF by utilizing biochemical, histopathological and molecular studies. RESULTS The UPLC analysis revealed the presence of 29.11% and 29.86% picroside I and II in the PF, respectively. In the zebrafish model of NASH, PF treatment reduced the hepatic lipid accumulation and modulated the expressions of lipogenic, inflammatory, oxidative, and cellular stress genes. Further, in MCD diet-induced NASH in mice, PF treatment showed a significant improvement in body weights and serum liver injury markers. Reduced degenerative changes and fibrous tissue was observed in the PF-treated groups. The downregulated expression of Srebp1c, Cd36, Fas, Chrebp, Pparγ, and Hnf4α showed anti-lipogenic potential of PF treatment. NASH development followed oxidative stress, mitochondrial dysfunction, and inflammation in the liver of mice. However, PF treatment encouraged mitochondrial biogenesis by upregulating Pgc1α, Tfam, and Nrf2 expressions. The elevated levels of NFκB, TNFα, IL6, TGFβ, and αSMA were also restored by PF, advocating its anti-inflammatory and anti-fibrogenic effect. CONCLUSION The present study revealed that PF ameliorate the progression of NASH by increasing mitochondrial biogenesis and decreasing lipogenesis, hepatic inflammation, and fibrosis.
Collapse
Affiliation(s)
- Swati Katoch
- Pharmacology and Toxicology Laboratory, Dietetics & Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, Himachal Pradesh 176061, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Jyoti Chhimwal
- Pharmacology and Toxicology Laboratory, Dietetics & Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, Himachal Pradesh 176061, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Damanpreet Singh
- Pharmacology and Toxicology Laboratory, Dietetics & Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, Himachal Pradesh 176061, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Dinesh Kumar
- Chemical Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, Himachal Pradesh 176061, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Vikram Patial
- Pharmacology and Toxicology Laboratory, Dietetics & Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, Himachal Pradesh 176061, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India.
| |
Collapse
|
35
|
Yoshimura Y, Matsui T, Kaneko N, Kobayashi I. Digestion and absorption of triacetin, a short-chain triacylglycerol. Lipids 2025. [PMID: 39891375 DOI: 10.1002/lipd.12433] [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: 08/17/2024] [Revised: 01/19/2025] [Accepted: 01/21/2025] [Indexed: 02/03/2025]
Abstract
Triacylglycerol (TG) is categorized into long-, medium-, and short-chain TG (SCTG). While the digestion of long- and medium-chain TG is well established, the process for SCTG remains unclear. This study investigated SCTG digestion by administering 2 mmol of triacetin to rats and analyzing acetin, acetic acid, and glycerol levels in the portal blood and small intestine. Triacetin was fully degraded in the upper gastrointestinal tract and absorbed as acetic acid and glycerol. Glycerol influx into the liver promoted gluconeogenesis, while acetate activated AMPK, resulting in the suppression of fatty acid synthesis-related genes and the upregulation of fatty acid β-oxidation-related genes. These findings demonstrate that triacetin not only serves as a substrate for energy metabolism but also regulates hepatic gene expression, highlighting its dual role as both a metabolic substrate and signaling molecule. Triacetin thus shows potential as a dietary modulator for improving metabolic health.
Collapse
Affiliation(s)
| | - Tomoka Matsui
- Department of Nutrition, Kobe Gakuin University, Kobe City, Japan
| | - Nagisa Kaneko
- Department of Nutrition, Kobe Gakuin University, Kobe City, Japan
| | - Ikuha Kobayashi
- Department of Nutrition, Kobe Gakuin University, Kobe City, Japan
| |
Collapse
|
36
|
Kim S, Han HJ, Rho H, Kang S, Mukherjee S, Kim J, Kim D, Ko HW, Lim SM, Im SS, Chung JY, Song J. Ebastine-mediated destabilization of E3 ligase MKRN1 protects against metabolic dysfunction-associated steatohepatitis. Cell Mol Life Sci 2025; 82:66. [PMID: 39888429 PMCID: PMC11785899 DOI: 10.1007/s00018-024-05535-2] [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: 06/20/2024] [Revised: 11/08/2024] [Accepted: 11/29/2024] [Indexed: 02/01/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a chronic condition encompassing metabolic dysfunction-associated steatotic liver (MASL) and metabolic dysfunction-associated steatohepatitis (MASH), which can progress to fibrosis, cirrhosis, or hepatocellular carcinoma (HCC). The heterogeneous and complex nature of MASLD complicates optimal drug development. Ebastine, an antihistamine, exhibits antitumor activity in various types of cancer. However, its effects on MASH remain unexplored. In the present study, we identified ebastine as a potential treatment for MASH. Our results indicated that ebastine acts as a novel MKRN1 inhibitor by promoting MKRN1 destabilization through self-ubiquitination, leading to AMP-activated protein kinase (AMPK) activation. Ebastine appeared to bind to the C-terminal domain of MKRN1, particularly at residues R298 and K360. Notably, Mkrn1 knockout (KO) mice demonstrated resistance to MASH, including obesity, steatosis, inflammation, and fibrosis under high-fat-high-fructose diet (HFHFD) conditions. Additionally, liver-specific Mkrn1 knockdown using AAV8 alleviated MASH symptoms in HFHFD-fed mice, implicating MKRN1 as a potential therapeutic target. Consistent with these findings, treatment with ebastine significantly reduced the risk of MASH in HFHFD-fed mice, with a decrease in MKRN1 expression and an increase in AMPK activity. Our study suggests that ebastine binds to MKRN1, promoting its destabilization and subsequent degradation by stimulating its ubiquitination. This enhances AMPK stability and activity, suppressing lipid accumulation, inflammation, and fibrosis. Moreover, the knockout of Mkrn1 mice decreased the risk of MASH, suggesting that ebastine could be a promising therapeutic agent for the treatment of MASH.
Collapse
Affiliation(s)
- Seungyeon Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Hyun-Ji Han
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Hyunjin Rho
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Subin Kang
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Sulagna Mukherjee
- Department of Physiology, Keimyung University School of Medicine, Daegu, 42601, Republic of Korea
| | - Jiwoo Kim
- Therapeutics & Biotechnology Division, Drug Discovery Platform Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon, 34114, Republic of Korea
| | - Doyoun Kim
- Therapeutics & Biotechnology Division, Drug Discovery Platform Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon, 34114, Republic of Korea
- Medicinal Chemistry and Pharmacology, Korea University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
- Brain Science Institute, Korea Institute of Science and Technology, Seoul, 07292, Republic of Korea
| | - Hyuk Wan Ko
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
- Department of Biochemistry, College of Life Science and Technology, Institute for Bio-medical Convergence Science and Technology, Yonsei University, Seoul, Republic of Korea
| | - Sang Min Lim
- Brain Science Institute, Korea Institute of Science and Technology, Seoul, 07292, Republic of Korea
| | - Seung-Soon Im
- Department of Physiology, Keimyung University School of Medicine, Daegu, 42601, Republic of Korea
| | - Joon-Yong Chung
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jaewhan Song
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea.
- Department of Biochemistry, College of Life Science and Technology, Institute for Bio-medical Convergence Science and Technology, Yonsei University, Seoul, Republic of Korea.
| |
Collapse
|
37
|
Cai F, Huang M, Liu W, Wan X, Qiu K, Xu X. Dietary addition of compound organic acids improves the growth performance, carcass trait, and body health of broilers. Front Nutr 2025; 12:1536606. [PMID: 39935581 PMCID: PMC11810740 DOI: 10.3389/fnut.2025.1536606] [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: 11/29/2024] [Accepted: 01/10/2025] [Indexed: 02/13/2025] Open
Abstract
Introduction The poultry industry constantly seeks strategies to enhance broiler growth performance and overall health. Organic acidifiers, including L-lactic acid, L-malic acid, and acetic acid, have gained attention as potential feed additives to improve animal production by modulating gut health, enhancing nutrient absorption, and supporting immune function. Despite their promising effects in other animal species, the impact of this novel compound organic acidifier on broiler performance, metabolism, and immune response has not been fully elucidated. This study aims to evaluate the effects of this compound acidifier on growth performance, serum lipid profile, antioxidant status, and immune parameters in broilers, providing insights into its potential benefits as a dietary supplement for broiler health and productivity. Methods A total of 240 broilers were randomly divided into four groups: a control group and three treatment groups receiving 0.25%, 0.5%, or 1.0% acidifier, with six replicates of ten birds each. Over a 6-week period, various parameters were measured, including serum triglycerides, high-density lipoproteins, lysozyme, immunoglobulins (IgA, IgM), superoxide dismutase (SOD) activity, IL-2, TNF-α, and gene expressions related to lipid metabolism. Results Over a 6-week period, the acidifier decreased serum triglycerides and high-density lipoproteins while also enhancing growth performance. Additionally, it raised the serum levels of lysozyme, IgA, IgM, and the SOD. Additionally, IL-2 and TNF-α concentrations in the jejunum mucosa decreased. The acidifier upregulated PPARα, AMPK, FABP1 and MTTP expressions, and downregulated APOB100. Overall, the acidifier effectively improved broiler growth performance during the early development phase primarily by enhancing hepatic lipid metabolism, antioxidant capacity, and immune function. Conclusion These results suggest that the acidifier may accelerate liver lipid metabolism in broilers by modulating the gene expression profiles involved in lipid metabolism.
Collapse
Affiliation(s)
- Fang Cai
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, China
| | - Meiping Huang
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, China
| | - Wei Liu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, China
| | - Xiaoling Wan
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, China
| | - Kai Qiu
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Xiao Xu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, China
| |
Collapse
|
38
|
Shi Q, Ran S, Song L, Yang H, Wang W, Liu H, Liu Q. NLRP6 overexpression improves nonalcoholic fatty liver disease by promoting lipid oxidation and decomposition in hepatocytes through the AMPK/CPT1A/PGC1A pathway. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2025; 45:118-125. [PMID: 39819720 PMCID: PMC11744278 DOI: 10.12122/j.issn.1673-4254.2025.01.15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Indexed: 01/19/2025]
Abstract
OBJECTIVES To investigate the regulatory role of nucleotide-bound oligomerized domain-like receptor containing pyrin-domain protein 6 (NLRP6) in liver lipid metabolism and non-alcoholic fatty liver disease (NAFLD). METHODS Mouse models with high-fat diet (HFD) feeding for 16 weeks (n=6) or with methionine choline-deficient diet (MCD) feeding for 8 weeks (n=6) were examined for the development of NAFLD using HE and oil red O staining, and hepatic expressions of NLRP6 were detected with RT-qPCR, Western blotting, and immunohistochemical staining. Cultured human hepatocytes (LO2 cells) with adenovirus-mediated NLRP6 overexpression or knock-down were treated with palmitic acid (PA) in the presence or absence of compound C (an AMPK inhibitor), and the changes in cellular lipid metabolism were examined by measuring triglyceride, ATP and β-hydroxybutyrate levels and using oil red staining, RT-qPCR, and Western blotting. RESULTS HFD and MCD feeding both resulted in the development of NAFLD in mice, which showed significantly decreased NLRP6 expression in the liver. In PA-treated LO2 cells, NLRP6 overexpression significantly decreased cellular TG content and lipid deposition, while NLRP6 knockdown caused the opposite effects. NLRP6 overexpression in PA-treated LO2 cells also increased mRNA and protein expressions of PGC1A and CPT1A, levels of ATP and β-hydroxybutyrate, and the phosphorylation level of AMPK pathway; the oxidative decomposition of lipids induced by Ad-NLRP6 was inhibited by the use of AMPK inhibitors. CONCLUSIONS NLRP6 overexpression promotes lipid oxidation and decomposition through AMPK/CPT1A/PGC1A to alleviate lipid deposition in hepatocytes.
Collapse
Affiliation(s)
- Qing Shi
- Department of Gastroenterology, Affiliated Hospital of Guizhou Medical University, Guiyang 550000, China
| | - Suye Ran
- Department of Gastroenterology, Affiliated Hospital of Guizhou Medical University, Guiyang 550000, China
| | - Lingyu Song
- Department of Gastroenterology, Affiliated Hospital of Guizhou Medical University, Guiyang 550000, China
| | - Hong Yang
- Department of Gastroenterology, Affiliated Hospital of Guizhou Medical University, Guiyang 550000, China
| | - Wenjuan Wang
- Department of Gastroenterology, Affiliated Hospital of Guizhou Medical University, Guiyang 550000, China
| | - Hanlin Liu
- Department of Gastroenterology, Affiliated Hospital of Guizhou Medical University, Guiyang 550000, China
| | - Qi Liu
- Department of Gastroenterology, Affiliated Hospital of Guizhou Medical University, Guiyang 550000, China
| |
Collapse
|
39
|
Li C, Wang F, Mao Y, Ma Y, Guo Y. Multi-omics reveals the mechanism of Trimethylamine N-oxide derived from gut microbiota inducing liver fatty of dairy cows. BMC Genomics 2025; 26:10. [PMID: 39762777 PMCID: PMC11702196 DOI: 10.1186/s12864-024-11067-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 11/19/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Trimethylamine N-oxide (TMAO) is a metabolite produced by gut microbiota, and its potential impact on lipid metabolism in mammals has garnered widespread attention in the scientific community. Bovine fatty liver disease, a metabolic disorder that severely affects the health and productivity of dairy cows, poses a significant economic burden on the global dairy industry. However, the specific role and pathogenesis of TMAO in bovine fatty liver disease remain unclear, limiting our understanding and treatment of the condition. This study aims to construct a bovine fatty liver cell model using an integrated approach that combines transcriptomic, proteomic, and metabolomic data. The objective is to investigate the impact of TMAO on lipid metabolism at the molecular level and explore its potential regulatory mechanisms. RESULTS We established an in vitro bovine fatty liver cell model and conducted a comprehensive analysis of cells treated with TMAO using high-throughput omics sequencing technologies. Bioinformatics methods were employed to delve into the regulatory effects on lipid metabolism, and several key genes were validated through RT-qPCR. Treatment with TMAO significantly affected 4790 genes, 397 proteins, and 137 metabolites. KEGG enrichment analysis revealed that the significantly altered molecules were primarily involved in pathways related to the pathology of fatty liver disease, such as metabolic pathways, insulin resistance, hepatitis B, and the AMPK signaling pathway. Moreover, through joint analysis, we further uncovered that the interaction between TMAO-mediated AMPK signaling and oxidative phosphorylation pathways might be a key mechanism promoting lipid accumulation in the liver. CONCLUSIONS Our study provides new insights into the role of TMAO in the pathogenesis of bovine fatty liver disease and offers a scientific basis for developing more effective treatment strategies.
Collapse
Affiliation(s)
- Chenlei Li
- College of Animal Science and Technology, Ningxia University, Yinchuan, 750021, China
- Key Laboratory of Ruminant Molecular and Cellular Breeding of Ningxia Hui Autonomous Region, College of Animal Science and Technology, Ningxia University, Yinchuan, 750021, China
| | - Feifei Wang
- College of Animal Science and Technology, Ningxia University, Yinchuan, 750021, China
- Key Laboratory of Ruminant Molecular and Cellular Breeding of Ningxia Hui Autonomous Region, College of Animal Science and Technology, Ningxia University, Yinchuan, 750021, China
| | - Yongxia Mao
- College of Animal Science and Technology, Ningxia University, Yinchuan, 750021, China
- Key Laboratory of Ruminant Molecular and Cellular Breeding of Ningxia Hui Autonomous Region, College of Animal Science and Technology, Ningxia University, Yinchuan, 750021, China
| | - Yanfen Ma
- College of Animal Science and Technology, Ningxia University, Yinchuan, 750021, China
- Key Laboratory of Ruminant Molecular and Cellular Breeding of Ningxia Hui Autonomous Region, College of Animal Science and Technology, Ningxia University, Yinchuan, 750021, China
| | - Yansheng Guo
- College of Animal Science and Technology, Ningxia University, Yinchuan, 750021, China.
- Key Laboratory of Ruminant Molecular and Cellular Breeding of Ningxia Hui Autonomous Region, College of Animal Science and Technology, Ningxia University, Yinchuan, 750021, China.
| |
Collapse
|
40
|
Bishr A, El-Mokadem BM, Gomaa AA. Canagliflozin alleviates acetaminophen-induced renal and hepatic injury in mice by modulating the p-GSK3β/Fyn-kinase/Nrf-2 and p-AMPK-α/STAT-3/SOCS-3 pathways. Sci Rep 2025; 15:729. [PMID: 39753621 PMCID: PMC11699121 DOI: 10.1038/s41598-024-82163-7] [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: 03/10/2024] [Accepted: 12/03/2024] [Indexed: 01/06/2025] Open
Abstract
Despite the fact that canagliflozin (Cana), a sodium-glucose cotransporter 2 inhibitor, is an anti-diabetic medication with additional effects on the kidney, there is limited experimental data to deliberate its hepato-reno-protective potentiality. Acetaminophen (APAP) overdose remains one of the prominent contributors to hepato-renal damage. AIM Our study assessed the novel effect of Cana against APAP-induced toxicities. MAIN METHODS mice were randomized into five groups: negative control, Cana25, APAP, Cana10 + APAP, and Cana25 + APAP. Cana was given for 5 days; a single dose of APAP was injected on the 6th day, followed by the scarification of animals 24 h later. KEY FINDINGS Pre-treatment with Cana ameliorated hepatic and renal functions, whereas, on the molecular levels, Cana promoted hepatic/renal P-AMP-activated protein kinase-α/ protein kinase B (p-Akt)/Glycogen synthase kinase (p-GSK3β) protein expression. Alternatively, Cana dampened the expression of STAT-3 and Fyn-kinase genes with a subsequent increase in the contents of suppressor of cytokine signaling (SOCS)-3 and also boosted the contents of the nuclear factor erythroid related factor 2 (Nrf-2)/heme oxygenase (HO)-1/ NADPH quinone oxidoreductase (NQO)-1 axis. The crosstalk between these paths ameliorated the APAP-induced hepatorenal structural alterations. SIGNIFICANCE Cana hepatorenal protective impact was provoked partly through modulating p-AMPK-α /SOCS-3/STAT-3 and GSK3β/Fyn-kinase signaling for its anti-inflammatory and antioxidant effects.
Collapse
Affiliation(s)
- Abeer Bishr
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ahram Canadian University, Giza, Egypt.
| | - Bassant M El-Mokadem
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian Chinese University, Cairo, Egypt
| | - Asmaa A Gomaa
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ahram Canadian University, Giza, Egypt
| |
Collapse
|
41
|
Singh LK, Pandey R, Siddiqi NJ, Sharma B. Molecular Mechanisms of Phthalate-Induced Hepatic Injury and Amelioration by Plant-Based Principles. TOXICS 2025; 13:32. [PMID: 39853030 PMCID: PMC11768991 DOI: 10.3390/toxics13010032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 12/24/2024] [Accepted: 12/26/2024] [Indexed: 01/26/2025]
Abstract
Phthalates are the emerging environmental toxicants derived from phthalic acid and its constituents, which are moderately present in plastics and many personal care products. Phthalate exposure occurs through various environmental factors, including air, water, and soil, with absorption facilitated via ingestion, inhalation, and dermal contact. Upon exposure, phthalates become bioavailable within the biological systems and undergo biotransformation and detoxification processes in the liver. The physicochemical properties of phthalates indicate their lipophilicity, environmental persistence, and bioaccumulation potential, influencing their absorption, distribution, and hepatic biotransformation. The prolonged exposure to phthalates adversely influences the biological redox system by altering the levels of the enzymatic and non-enzymatic antioxidants, molecular signaling pathways, and causing hepatic pathogenesis. The strategies to combat phthalate-induced toxicity include avoiding exposure to these compounds and using plant-based bioactive molecules such as polyphenols, which possess therapeutic potential as antioxidants, suppress inflammatory cascades, prevent oxidative damage, and stabilize cellular integrity. This review presents a comprehensive and updated account of the chemical, biochemical, immunological, and toxicological properties of phthalates, along with novel plant-based therapeutic strategies to mitigate the phthalate-induced adverse effects on living systems.
Collapse
Affiliation(s)
- Lalit Kumar Singh
- Department of Biochemistry, University of Allahabad, Prayagraj 211002, Uttar Pradesh, India;
| | - Rashmi Pandey
- Department of Biochemistry, Government Medical College, Haridwar 247667, Uttarakhand, India
| | - Nikhat Jamal Siddiqi
- Department of Internal Surgical Nursing, College of Nursing, King Saud University, Riyadh 11421, Saudi Arabia
| | - Bechan Sharma
- Department of Biochemistry, University of Allahabad, Prayagraj 211002, Uttar Pradesh, India;
| |
Collapse
|
42
|
Tang M, Wang X, Wang S, Xing C, Xu Q, Mu Y, Wu X, Zhao ZA, Li F. 10-Hydroxy-2-decenoic acid attenuates nonalcoholic fatty liver disease by activating AMPK-α signaling pathway. Biochem Pharmacol 2025; 231:116648. [PMID: 39581533 DOI: 10.1016/j.bcp.2024.116648] [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: 06/23/2024] [Revised: 11/01/2024] [Accepted: 11/21/2024] [Indexed: 11/26/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD) originates from metabolic dysfunctions, is one of the most commonly encountered liver disorders worldwide, characterized by ectopic lipid deposition within hepatocytes, accompanied by hepatocellular injury and necroinflammation. Currently, NAFLD has very few treatment options. Purified from royal jelly, 10-hydroxy-2-decenoic acid (10-HDA) is the primary bioactive ingredient with a series of beneficial effects against various metabolic diseases. Herein, we investigated the effects of 10-HDA in methionine and choline deficiency (MCD) diet induced NAFLD model and free fatty acids (FFAs) induced lipid-laden hepatocyte model and explored the underlying mechanisms. In the mice fed with MCD diet, 10-HDA treatment significantly reduced hepatic steatosis, hepatocellular injury, apoptosis, inflammatory response and fibrosis. In vitro, 10-HDA treatment reduced lipid accumulation and apoptosis in hepatocytes induced by FFAs. Mechanistically, 10-HDA therapy restored AMPK-α phosphorylation, leading to the phosphorylation and inactivation acetyl-CoA carboxylase (ACC). Consequently, this increased the expression of carnitine palmitoyl transferase 1α(CPT1α), and peroxisome proliferators-activated receptors α (PPARα), and lowered the expression of cleavage forms of sterol regulatory element binding protein-1 (SREBP-1) and fatty acid synthetase (FASN). Furthermore, pretreating the cells with the AMPK-α inhibitor, compound C, greatly eliminated these beneficial effects of 10-HDA. Additionally, molecular docking analysis indicated that 10-HDA bound the domain of AMPK-α1 subunit. Based on these findings, 10-HDA suppresses hepatic lipogenesis via AMPK-α-dependent suppression of the ACC pathway, thus inhibiting hepatocellular injury, apoptosis, inflammatory response and fibrosis. 10-HDA may represent a promising candidate drug for the treatment of NAFLD.
Collapse
Affiliation(s)
- Minyi Tang
- The School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Xinzi Wang
- The School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Shuai Wang
- The School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Chaofeng Xing
- Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Southern Medical University, Foshan 528308, China
| | - Qihua Xu
- Bioscience and Biomedical Engineering Thrust, The Hong Kong University of Science and Technology (Guangzhou), Guangzhou 511400, China
| | - Yunping Mu
- The School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Xiaoli Wu
- The School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China.
| | - Zijian Allan Zhao
- Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Southern Medical University, Foshan 528308, China.
| | - Fanghong Li
- The School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China.
| |
Collapse
|
43
|
Jiang J, Sun C, Wang G, Xu Q, Bian Y, Li J, Li J, Ding R, Lin H, Tian W, Chen H. C-13 Norisoprenoids and Eudesmanoids from Nelumbo nucifera Gaertn. Regulate the Lipid Metabolism via the AMPK/ACC/SREBP-1c Signaling Pathway. Chem Biodivers 2025; 22:e202401778. [PMID: 39474994 DOI: 10.1002/cbdv.202401778] [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: 07/22/2024] [Accepted: 09/04/2024] [Indexed: 01/19/2025]
Abstract
Lotustine A (1), an undescribed C-13 norisoprenoid, along with 22 known analogues and two eudesmanoids, were isolated from the aerial parts of Nelumbo nucifera Gaertn. Among them, compounds 2, 15, 17, 21, 22, 24, 25 were isolated from N. nucifera leaves for the first time. Their structures, including absolute configurations, were elucidated by nuclear magnetic resonance, mass spectroscopy, and the modified Mosher's method. Compound 1 is the first example of C-13 norisoprenoid with a terminal double bond between C-5 and C-13. Moreover, the lipid-lowering activities of the isolates were evaluated, and the results showed that 2, 24 and 25 could remarkably decrease the levels of both total cholesterol and triglyceride in free fatty acids induced HepG2 cells at the concentration of 20 μM. The oil red staining assay further demonstrated the lipid-lowering effects of 2, 24 and 25. The western blot results indicated that compounds 2, 24 and 25 could regulate the lipid metabolism via the activation of the AMPK/ACC/SREBP-1c signaling pathway.
Collapse
Affiliation(s)
- Jian Jiang
- Chen Fujian Provincial Key Laboratory of Innovative Drug Target, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, People's Republic of China
| | - Cuiling Sun
- Chen Fujian Provincial Key Laboratory of Innovative Drug Target, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, People's Republic of China
| | - Guanghui Wang
- Chen Fujian Provincial Key Laboratory of Innovative Drug Target, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, People's Republic of China
| | - Qinnan Xu
- Chen Fujian Provincial Key Laboratory of Innovative Drug Target, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, People's Republic of China
| | - Yuting Bian
- Chen Fujian Provincial Key Laboratory of Innovative Drug Target, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, People's Republic of China
| | - Jie Li
- Chen Fujian Provincial Key Laboratory of Innovative Drug Target, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, People's Republic of China
| | - Jingdian Li
- Chen Fujian Provincial Key Laboratory of Innovative Drug Target, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, People's Republic of China
| | - Rong Ding
- Chen Fujian Provincial Key Laboratory of Innovative Drug Target, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, People's Republic of China
| | - Houwen Lin
- Chen Fujian Provincial Key Laboratory of Innovative Drug Target, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, People's Republic of China
- State Key Laboratory of Oncogene and Related Genes, Department of Pharmacy, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, People's Republic of China
| | - Wenjing Tian
- Chen Fujian Provincial Key Laboratory of Innovative Drug Target, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, People's Republic of China
| | - Haifeng Chen
- Chen Fujian Provincial Key Laboratory of Innovative Drug Target, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, People's Republic of China
| |
Collapse
|
44
|
Hamamah S, Iatcu OC, Covasa M. Dietary Influences on Gut Microbiota and Their Role in Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD). Nutrients 2024; 17:143. [PMID: 39796579 PMCID: PMC11722922 DOI: 10.3390/nu17010143] [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: 12/15/2024] [Revised: 12/27/2024] [Accepted: 12/30/2024] [Indexed: 01/13/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a major contributor to liver-related morbidity, cardiovascular disease, and metabolic complications. Lifestyle interventions, including diet and exercise, are first line in treating MASLD. Dietary approaches such as the low-glycemic-index Mediterranean diet, the ketogenic diet, intermittent fasting, and high fiber diets have demonstrated potential in addressing the metabolic dysfunction underlying this condition. The development and progression of MASLD are closely associated with taxonomic shifts in gut microbial communities, a relationship well-documented in the literature. Given the importance of diet as a primary treatment for MASLD, it is important to understand how gut microbiota and their metabolic byproducts mediate favorable outcomes induced by healthy dietary patterns. Conversely, microbiota changes conferred by unhealthy dietary patterns such as the Western diet may induce dysbiosis and influence steatotic liver disease through promoting hepatic inflammation, up-regulating lipogenesis, dysregulating bile acid metabolism, increasing insulin resistance, and causing oxidative damage in hepatocytes. Although emerging evidence has identified links between diet, microbiota, and development of MASLD, significant gaps remain in understanding specific microbial roles, metabolite pathways, host interactions, and causal relationships. Therefore, this review aims to provide mechanistic insights into the role of microbiota-mediated processes through the analysis of both healthy and unhealthy dietary patterns and their contribution to MASLD pathophysiology. By better elucidating the interplay between dietary nutrients, microbiota-mediated processes, and the onset and progression of steatotic liver disease, this work aims to identify new opportunities for targeted dietary interventions to treat MASLD efficiently.
Collapse
Affiliation(s)
- Sevag Hamamah
- Department of Internal Medicine, Scripps Mercy Hospital, San Diego, CA 92103, USA;
| | - Oana C. Iatcu
- Department of Biomedical Sciences, College of Medicine and Biological Science, University of Suceava, 720229 Suceava, Romania;
| | - Mihai Covasa
- Department of Biomedical Sciences, College of Medicine and Biological Science, University of Suceava, 720229 Suceava, Romania;
| |
Collapse
|
45
|
Cheng H, Wu J, Peng H, Li J, Liu Z, Wang X, Zhang K, Xie L. Epigenetic Modulation with 5-Aza-CdR Prevents Metabolic-Associated Fatty Liver Disease Promoted by Maternal Overnutrition. Nutrients 2024; 17:106. [PMID: 39796540 PMCID: PMC11722594 DOI: 10.3390/nu17010106] [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: 10/28/2024] [Revised: 12/03/2024] [Accepted: 12/12/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND/OBJECTIVES This study builds on previous findings from mouse models, which showed that maternal overnutrition induced by a high-fat diet (HFD) promotes metabolic-associated fatty liver disease (MAFLD) in offspring, linked to global DNA hypermethylation. We explored whether epigenetic modulation with 5-Aza-CdR, a DNA methylation inhibitor, could prevent MAFLD in offspring exposed to maternal overnutrition. METHODS The offspring mice from dams of maternal overnutrition were fed either a chow diet or a high-fat diet (HFD) for 10 weeks. These mice were randomly divided into two groups: HFD, and AZA + HFD. Mice assigned to the AZA group were given 5-Aza-CdR during the last three weeks. RESULTS Our findings show that 5-Aza-CdR treatment in HFD-fed offspring effectively countered weight gain, improved glucose regulation, and minimized hepatic fat buildup along with serum lipid imbalances. Additionally, it boosted AMPK signaling and raised PPAR-α expression, pointing to enhanced fatty acid oxidation. We also detected an increase in JNK signaling, affecting the gene expression associated with cell death and proliferation. Notably, treated mice displayed more hepatic inflammation than the HFD group alone, suggesting a complex, dual impact on MAFLD management. Significant apoptotic and inflammatory gene changes were identified, along with corresponding differentially methylated regions triggered by 5-Aza-CdR, marking potential therapeutic targets. CONCLUSIONS 5-Aza-CdR was shown to mitigate MAFLD features in offspring of maternal overnutrition by reversing DNA hypermethylation and improving metabolic pathways, though its dual impact on inflammation highlights the need for further research to optimize its therapeutic potential.
Collapse
Affiliation(s)
- Henghui Cheng
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA; (H.C.); (H.P.); (Z.L.); (X.W.)
| | - Jie Wu
- Center for Epigenetics & Disease Prevention, Institute of Biosciences & Technology, College of Medicine, Texas A&M University, Houston, TX 77030, USA; (J.W.); (J.L.)
| | - Hui Peng
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA; (H.C.); (H.P.); (Z.L.); (X.W.)
| | - Jiangyuan Li
- Center for Epigenetics & Disease Prevention, Institute of Biosciences & Technology, College of Medicine, Texas A&M University, Houston, TX 77030, USA; (J.W.); (J.L.)
- Department of Statistics, Texas A&M University, College Station, TX 77843, USA
| | - Zhimin Liu
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA; (H.C.); (H.P.); (Z.L.); (X.W.)
| | - Xian Wang
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA; (H.C.); (H.P.); (Z.L.); (X.W.)
| | - Ke Zhang
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA; (H.C.); (H.P.); (Z.L.); (X.W.)
- Center for Epigenetics & Disease Prevention, Institute of Biosciences & Technology, College of Medicine, Texas A&M University, Houston, TX 77030, USA; (J.W.); (J.L.)
| | - Linglin Xie
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA; (H.C.); (H.P.); (Z.L.); (X.W.)
| |
Collapse
|
46
|
Elshaer A, Lizaola-Mayo BC. Evaluating the Role of Aspirin in Liver Disease: Efficacy, Safety, Potential Benefits and Risks. Life (Basel) 2024; 14:1701. [PMID: 39768407 PMCID: PMC11679757 DOI: 10.3390/life14121701] [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: 11/07/2024] [Revised: 12/15/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025] Open
Abstract
The rise in liver disease incidence and prevalence has led to increasing morbidity and mortality worldwide. Persistent hepatic inflammation drives disease progression by increasing fibrosis, advancing to cirrhosis, and potentially developing into hepatocellular carcinoma (HCC). Addressing these complications is essential to reduce liver-related mortality. Recent studies suggest that non-steroidal anti-inflammatory drugs, particularly aspirin, may play a beneficial role in managing liver disease. Aspirin's anti-inflammatory and chemoprotective effects contribute to slowing disease progression and reducing the risks associated with chronic liver disease (CLD). This review highlights the current literature on the effects of aspirin in CLD, with a focus on patients with metabolic-associated steatotic liver disease (MASLD) and hepatitis B and C. We will examine aspirin's potential ability to mitigate fibrosis, reduce the incidence of HCC, and lower liver-related mortality. Additionally, we will discuss its potential side effects and safety considerations, particularly in the context of liver disease, where there is an increased risk of bleeding.
Collapse
Affiliation(s)
- Amani Elshaer
- Department of Internal Medicine, Mayo Clinic, Phoenix, AZ 85054, USA;
| | - Blanca C. Lizaola-Mayo
- Division of Gastroenterology, Hepatology and Transplant Hepatology, Mayo Clinic, Phoenix, AZ 85054, USA
| |
Collapse
|
47
|
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
|
48
|
Qu K, Shi M, Chen L, Liu Y, Yao X, Li X, Tan B, Xie S. Residual levels of dietary deltamethrin interfere with growth and intestinal health in Litopenaeus vannamei. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 288:117376. [PMID: 39612679 DOI: 10.1016/j.ecoenv.2024.117376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 10/24/2024] [Accepted: 11/18/2024] [Indexed: 12/01/2024]
Abstract
To date, few study explored the damage of chronic dietary exposure to the lipophilic pesticide deltamethrin (DM) in aquatic animals, and it remains unclear whether its toxicity and residue levels would be affected by dietary lipid levels. Therefore, the present study aimed to elucidate the interactions between dietary lipid levels and DM levels in the Pacific white shrimp, focusing on growth performance, antioxidant capacity, and intestinal microbiota. DM has excellent insecticidal activity and has been used worldwide. Previous research has shown that environmental DM poses toxicity risks to aquatic animals. Six different diets were formulated to feed shrimp for 6 weeks with two lipid levels (6.96 %, 10.88 %) and three DM levels (0.2 mg·kg-1, 1 mg·kg-1, 5 mg·kg-1), namely LF0.2, LF1, LF5, HF0.2, HF1, HF5, respectively. Each diet was assigned to three net cages with a total of 18 cages (40 shrimp per tank, average weight (0.382±0.001 g), of which 0.2 mg·kg-1, are grouped in environmental DM control groups. The growth of shrimp was reduced as the dietary DM levels increased. When shrimp were fed a diet containing a high dose of DM, a reduction in their antioxidant capacity was also observed. Enzyme activity and gene expression related to lipid metabolism in hepatopancreas and hemolymph indicated a significant interaction between dietary lipid levels and DM in the lipid metabolism of shrimp. The terms of detoxification-related genes (gst, sult, cyp1a1) were upregulated in shrimp fed the high-dose DM. Additionally, the presence of DM in the diet severely harmed the hepatopancreas and intestinal histological morphology. DM in the diet increased the susceptibility of shrimp to pathogens and induced intestine microbiota dysbiosis, disrupting the balance of inter-species interactions. DM was not detected in the muscle and hepatopancreas of the shrimp after six weeks of exposure. In conclusion, the presence of DM in feed reduced the growth performance and antioxidant capacity of shrimp, damaging intestinal health. DM was rapidly metabolized by shrimp.
Collapse
Affiliation(s)
- Kangyuan Qu
- College of Aquatic Sciences, Guangdong Ocean University, Zhanjiang 524088, China
| | - Menglin Shi
- College of Aquatic Sciences, Guangdong Ocean University, Zhanjiang 524088, China
| | - Liutong Chen
- College of Aquatic Sciences, Guangdong Ocean University, Zhanjiang 524088, China
| | - Yucheng Liu
- College of Aquatic Sciences, Guangdong Ocean University, Zhanjiang 524088, China
| | - Xinzhou Yao
- College of Aquatic Sciences, Guangdong Ocean University, Zhanjiang 524088, China
| | - Xiaoyue Li
- College of Aquatic Sciences, Guangdong Ocean University, Zhanjiang 524088, China
| | - Beiping Tan
- College of Aquatic Sciences, Guangdong Ocean University, Zhanjiang 524088, China; Guangdong Province Research Center for Accurate Nutrition and High-Efficiency Feeding of Aquatic Animals, Zhanjiang 524088, China; Key Laboratory of Aquatic Feed Science and Technology for Livestock and Poultry in Southern China, under the Ministry of Agriculture, Zhanjiang 524088, China
| | - Shiwei Xie
- College of Aquatic Sciences, Guangdong Ocean University, Zhanjiang 524088, China; Guangdong Province Research Center for Accurate Nutrition and High-Efficiency Feeding of Aquatic Animals, Zhanjiang 524088, China; Key Laboratory of Aquatic Feed Science and Technology for Livestock and Poultry in Southern China, under the Ministry of Agriculture, Zhanjiang 524088, China.
| |
Collapse
|
49
|
Geng R, Guo J, Lao Y, Kang SG, Huang K, Tong T. Chronic UVB exposure induces hepatic injury in mice: Mechanistic insights from integrated multi-omics. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 362:124933. [PMID: 39265770 DOI: 10.1016/j.envpol.2024.124933] [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: 06/08/2024] [Revised: 07/28/2024] [Accepted: 09/08/2024] [Indexed: 09/14/2024]
Abstract
Chronic UVB exposure poses a significant threat to both skin and visceral health. In recent years, the adverse role of chronic UVB exposure in liver health has been suggested but not fully elucidated. This study aims to comprehensively investigate the effects of chronic UVB exposure on liver health in male SKH-1 hairless mice and clarify potential mechanisms through multi-omics approaches. The findings suggested that 10-week chronic skin exposure to UVB not only triggers hepatic inflammation and oxidative stress but also, more importantly, results in lipid metabolism abnormalities in the liver. Hepatic transcriptomic analysis revealed significant alterations in various signaling pathways and physiological processes associated with inflammation, oxidative stress, and lipid metabolism. Further lipidomic analysis illustrated significant changes in the metabolism of glycerolipids, sphingolipids, and glycerophospholipids in the liver following chronic UVB exposure. The 16S rRNA sequencing analysis indicated that chronic UVB exposure disrupts the structure and function of the microbiota. In search of potential mechanisms used by the microbiome to regulate the hepatic disease morphology, we filtered mouse fecal supernatants and cultured the supernatants with HepG2 cells. Fecal supernatant from UVB-exposed mice induced increased secretion of the inflammatory cytokine IL-8, accumulation of MDA, reduced SOD activity, and decreased lipid content in normal hepatic cells. In summary, skin chronic exposure to UVB induces multiple liver injuries and gut microbiota dysbiosis in mice and gut microbiota metabolites may be one of the contributing factors to hepatic injury caused by chronic UVB exposure. These discoveries deepen the comprehension of the health risks associated with chronic UVB exposure.
Collapse
Affiliation(s)
- Ruixuan Geng
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China; Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing, 100083, China; Beijing Laboratory for Food Quality and Safety, Beijing, 100083, China
| | - Jingya Guo
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China; Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing, 100083, China; Beijing Laboratory for Food Quality and Safety, Beijing, 100083, China
| | - Yujie Lao
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China; Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing, 100083, China; Beijing Laboratory for Food Quality and Safety, Beijing, 100083, China
| | - Seong-Gook Kang
- Department of Food Engineering and Solar Salt Research Center, Mokpo National University, Muangun, 58554, Republic of Korea
| | - Kunlun Huang
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China; Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing, 100083, China; Beijing Laboratory for Food Quality and Safety, Beijing, 100083, China
| | - Tao Tong
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China; Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing, 100083, China; Beijing Laboratory for Food Quality and Safety, Beijing, 100083, China.
| |
Collapse
|
50
|
Wan W, Wei R, Xu B, Cao H, Zhi Y, Guo F, Liu H, Li B, Wu J, Gao Y, Zhang K. Qiwei Jinggan Ling regulates oxidative stress and lipid metabolism in alcoholic liver disease by activating AMPK. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156125. [PMID: 39388920 DOI: 10.1016/j.phymed.2024.156125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/24/2024] [Accepted: 07/09/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND Alcoholic liver disease (ALD) is a severe public health concern worldwide and there is still a lack of effective treatments. Qiwei Jinggan Ling (QJL) has protective effects against various liver injuries, but its pharmacological action on ALD has received little attention. PURPOSE To investigate the effect and mechanism of QJL on ALD in vivo and in vitro. METHODS In vivo, an ALD mouse model was established by alcohol combined with a high-fat diet (HFD) and treated with QJL. Biochemical indicators, HE staining, and Oil Red O staining were employed to assess hepatic oxidative stress, steatosis, and alcohol metabolism. RNA sequencing analysis was performed, and the results were verified by qRT-PCR and Western blot to elucidate the hepatoprotective mechanism of QJL. In vitro, HepG2 cells were co-stimulated with NaOA (sodium oleate) and EtOH (ethanol), followed by intervention with Compound C (CC, AMPK inhibitor) and QJL-containing serum. Oil Red O, BODIPY (boron-dipyrromethene), and ROS (reactive oxygen species) staining were applied to validate the efficacy and mechanism of QJL-containing serum. The expression of AMP-activated protein kinase (AMPK) pathway-related factors was analyzed through qRT-PCR and Western blot for additional corroboration. Moreover, the key pharmacodynamic components of QJL were identified by UPLC-MS/MS and molecular docking. RESULTS In vivo, QJL ameliorated liver structural disorders, steatosis, oxidative stress, and impaired alcohol metabolism, as indicated by biochemical indicators and histopathological assays. RNA sequencing analysis revealed that QJL reversed the expression of genes related to alcohol metabolism, fatty acid metabolism, and cholesterol metabolism. The results of qRT-PCR and Western blot were in line with those of RNA sequencing. Furthermore, it was discovered that QJL significantly upregulated the expression of p-AMPK and downregulated the expression of sterol regulatory element binding transcription factor 1 (SREBP-1c). In vitro, biochemical indicators and staining assays demonstrated that QJL-containing serum inhibited lipid accumulation and oxidative stress. The qRT-PCR and Western blot analysis revealed that QJL-containing serum markedly enhanced the expression of p-AMPK and carnitine palmitoyltransferase 1a (Cpt1a), while suppressing the expression of SREBP-1c, fatty acid synthase (Fasn), and acetyl-coenzyme A carboxylase 1 (ACC-1). However, CC inhibited the above pharmacological activities of QJL-containing serum. Additionally, (2S)-Liquiritigenin, Glycyrrhetinate, Isovitexin, Taxifolin, and Yohimbine were proved to be the key active components of QJL. CONCLUSION QJL had the potential to be a therapeutic drug for ALD by activating the AMPK pathway, thereby regulating lipid metabolism and inhibiting oxidative stress.
Collapse
Affiliation(s)
- Weimin Wan
- Pharmacology Laboratory of Prevention and Treatment of High Incidence of Disease, Guilin Medical University, Guilin 541199, Guangxi, China
| | - Riming Wei
- Pharmacology Laboratory of Prevention and Treatment of High Incidence of Disease, Guilin Medical University, Guilin 541199, Guangxi, China
| | - Baoling Xu
- Pharmacology Laboratory of Prevention and Treatment of High Incidence of Disease, Guilin Medical University, Guilin 541199, Guangxi, China; Department of Emergency, The Second Affiliated Hospital of Guilin Medical University, Guilin 541199, Guangxi, China
| | - Houkang Cao
- Pharmacology Laboratory of Prevention and Treatment of High Incidence of Disease, Guilin Medical University, Guilin 541199, Guangxi, China
| | - Yueping Zhi
- Pharmacology Laboratory of Prevention and Treatment of High Incidence of Disease, Guilin Medical University, Guilin 541199, Guangxi, China
| | - Fengyue Guo
- Pharmacology Laboratory of Prevention and Treatment of High Incidence of Disease, Guilin Medical University, Guilin 541199, Guangxi, China
| | - Haiping Liu
- Pharmacology Laboratory of Prevention and Treatment of High Incidence of Disease, Guilin Medical University, Guilin 541199, Guangxi, China
| | - Bo Li
- Pharmacology Laboratory of Prevention and Treatment of High Incidence of Disease, Guilin Medical University, Guilin 541199, Guangxi, China
| | - Jianzhao Wu
- Pharmacology Laboratory of Prevention and Treatment of High Incidence of Disease, Guilin Medical University, Guilin 541199, Guangxi, China
| | - Ya Gao
- Pharmacology Laboratory of Prevention and Treatment of High Incidence of Disease, Guilin Medical University, Guilin 541199, Guangxi, China
| | - Kefeng Zhang
- Pharmacology Laboratory of Prevention and Treatment of High Incidence of Disease, Guilin Medical University, Guilin 541199, Guangxi, China.
| |
Collapse
|