1
|
Li Z, Chen Y, Shi T, Cao H, Chen G, Yu L. Potential of queen bee larvae as a dietary supplement for obesity management: modulating the gut microbiota and promoting liver lipid metabolism. Food Funct 2025; 16:3848-3861. [PMID: 40131738 DOI: 10.1039/d5fo00166h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2025]
Abstract
Queen bee larvae (QBL) have been consumed as both a traditional food and medicine in China for thousands of years; however, their specific benefits for human health, particularly their potential anti-obesity property, remain underexplored. This study investigated the anti-obesity effect of QBL freeze-dried powder (QBLF) on high-fat diet (HFD) induced obesity in mice and explored the underlying mechanisms. Our findings showed that QBLF effectively reduced body weight, fasting blood glucose levels, lipid accumulation, and inflammation in HFD mice. 16S rRNA sequencing revealed that QBLF significantly modulated the gut microbiota disrupted by an HFD, notably increasing the relative abundance of beneficial microbes such as Ileibacterium, Clostridium sensu stricto 1, Incertae sedis, Streptococcus, Lactococcus, Clostridia UCG-014, and Lachnospiraceae UCG-006, which were inversely associated with obesity-related phenotypes in the mice. RNA sequencing analysis further demonstrated that QBLF intervention upregulated the expression of genes involved in liver lipid metabolism, including Pck1, Cyp4a10, Cyp4a14, and G6pc, while downregulating genes associated with the inflammatory response, such as Cxcl10, Ccl2, Traf1, Mapk15, Lcn2, and Fosb. These results suggested that QBLF can ameliorate HFD-induced obesity through regulating the gut microbiota, promoting liver lipid metabolism, and reducing inflammatory response.
Collapse
Affiliation(s)
- Zhuang Li
- School of Plant Protection, Anhui Province Key Laboratory of Crop Integrated Pest Management, Hefei 230031, China.
- Apiculture Research Institute, Anhui Agricultural University, Hefei 230031, China
- Biotechnology Center of Anhui Agriculture University, Hefei 230031, China
| | - Yiang Chen
- National Key Laboratory for Tea Plant Germplasm Innovation and Resource Utilization, School of Tea Science, Anhui Agricultural University, Hefei, 230036, China.
| | - Tengfei Shi
- School of Plant Protection, Anhui Province Key Laboratory of Crop Integrated Pest Management, Hefei 230031, China.
- Apiculture Research Institute, Anhui Agricultural University, Hefei 230031, China
- Biotechnology Center of Anhui Agriculture University, Hefei 230031, China
| | - Haiqun Cao
- School of Plant Protection, Anhui Province Key Laboratory of Crop Integrated Pest Management, Hefei 230031, China.
- Apiculture Research Institute, Anhui Agricultural University, Hefei 230031, China
- Biotechnology Center of Anhui Agriculture University, Hefei 230031, China
| | - Guijie Chen
- National Key Laboratory for Tea Plant Germplasm Innovation and Resource Utilization, School of Tea Science, Anhui Agricultural University, Hefei, 230036, China.
| | - Linsheng Yu
- School of Plant Protection, Anhui Province Key Laboratory of Crop Integrated Pest Management, Hefei 230031, China.
- Apiculture Research Institute, Anhui Agricultural University, Hefei 230031, China
- Biotechnology Center of Anhui Agriculture University, Hefei 230031, China
| |
Collapse
|
2
|
Zhu Q, Xie X, Fang L, Huang C, Li J. Chronic alcohol intake disrupts cytochrome P450 enzyme activity in alcoholic fatty liver disease: insights into metabolic alterations and therapeutic targets. Front Chem 2025; 13:1509785. [PMID: 40433307 PMCID: PMC12106329 DOI: 10.3389/fchem.2025.1509785] [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: 10/11/2024] [Accepted: 01/08/2025] [Indexed: 05/29/2025] Open
Abstract
Introduction Alcoholic fatty liver disease (AFLD) is a common consequence of chronic alcohol consumption, characterized by lipid accumulation and oxidative stress in the liver. Cytochrome P450 (CYP450) enzymes play essential roles in metabolizing alcohol and other compounds. However, the specific long-term effects of alcohol on these enzymes remain unclear. Methods This study the examines influence of prolonged ethanol exposure on CYP450 activity and expression in AFLD using a rat model. Key enzymes such as CYP2E1, CYP2D6, and CYP3A1 were assessed in relation to lipid accumulation and oxidative stress. Results Significant alterations were identified in the expression and activity of CYP2E1, CYP2D6, and CYP3A1, which were associated with increased lipid accumulation and oxidative stress in the liver. Additionally, the expression of P-glycoprotein (P-gp) was elevated, suggesting that chronic alcohol intake may impact drug transport and excretion. Discussion These findings provide new insights into the molecular mechanisms of AFLD and highlight the potential of CYP450 modulation as a therapeutic target. By elucidating how long-term ethanol exposure disrupts hepatic CYP450 enzyme profiles, this research lays the groundwork for developing personalized therapeutic strategies to improve outcomes for patients with AFLD.
Collapse
Affiliation(s)
- Qian Zhu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Xuefeng Xie
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Ling Fang
- The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Cheng Huang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Jun Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| |
Collapse
|
3
|
Xu K, Corona-Avila I, Frutos MD, Núñez-Sánchez MÁ, Makhanasa D, Shah PV, Guzman G, Ramos-Molina B, Priyadarshini M, Khan MW. Hepatic HKDC1 deletion alleviates western diet-induced MASH in mice. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167746. [PMID: 40020530 DOI: 10.1016/j.bbadis.2025.167746] [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: 12/02/2024] [Revised: 02/05/2025] [Accepted: 02/20/2025] [Indexed: 03/03/2025]
Abstract
The global prevalence of Metabolic Dysfunction-Associated Steatohepatitis (MASH) has been rising sharply, closely mirroring the increasing rates of obesity and metabolic syndrome. MASH exhibits a strong sexual dimorphism where females are affected with more severe forms after menopause. Hexokinase domain-containing protein 1 (HKDC1) has recently been recognized for its role in liver diseases, where its expression is minimal under normal conditions but significantly increases in response to metabolic stressors like obesity and liver injury. This selective upregulation suggests HKDC1's potential specialization in hepatic glucose and lipid dysregulation, linking it closely to the progression of MASH. This study aims to clarify the role of HKDC1 in Western diet-induced MASH in female mice by examining its impact on hepatic glucose and lipid metabolism, offering insights into its potential as a therapeutic target and addressing the need for sex-specific research in liver disease. This study reveals that HKDC1 expression is elevated in obese women with MASH and correlates with liver pathology. In a mouse model, liver-specific HKDC1 knockout (HKDC1LKO) protected against Western diet-induced obesity, glucose intolerance, and MASH features, including steatosis, inflammation, and fibrosis. Transcriptomic analysis showed that HKDC1 deletion reduced pro-inflammatory and pro-fibrotic gene expression, while gut microbiome analysis indicated a shift toward MASH-protective bacteria. These findings suggest that HKDC1 may exacerbate MASH progression through its role in metabolic and inflammatory pathways, making it a potential therapeutic target.
Collapse
Affiliation(s)
- Kai Xu
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Illinois at Chicago, Chicago, United States of America
| | - Irene Corona-Avila
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Illinois at Chicago, Chicago, United States of America
| | - María Dolores Frutos
- Department of General and Digestive System Surgery, Virgen de la Arrixaca University Hospital, 30120 Murcia, Spain
| | - María Ángeles Núñez-Sánchez
- Obesity, Diabetes and Metabolism Research Group, Biomedical Research Institute of Murcia (IMIB), 30120 Murcia, Spain
| | - Dhruvi Makhanasa
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Illinois at Chicago, Chicago, United States of America
| | - Pratham Viral Shah
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Illinois at Chicago, Chicago, United States of America
| | - Grace Guzman
- Department of Pathology, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Bruno Ramos-Molina
- Obesity, Diabetes and Metabolism Research Group, Biomedical Research Institute of Murcia (IMIB), 30120 Murcia, Spain
| | - Medha Priyadarshini
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Illinois at Chicago, Chicago, United States of America.
| | - Md Wasim Khan
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Illinois at Chicago, Chicago, United States of America.
| |
Collapse
|
4
|
Huda N, Kusumanchi P, Jiang Y, Gao H, Thoudam T, Zeng G, Skill NJ, Sun Z, Liangpunsakul S, Ma J, Yang Z. Silencing FAF2 mitigates alcohol-induced hepatic steatosis by modulating lipolysis and PCSK9 pathway. Hepatol Commun 2025; 9:e0641. [PMID: 39969435 PMCID: PMC11841855 DOI: 10.1097/hc9.0000000000000641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 12/02/2024] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Chronic alcohol consumption leads to lipid accumulation, oxidative stress, cellular damage, and inflammation in the liver, collectively referred to as alcohol-associated liver disease (ALD). FAF2/UBXD8/ETEA (Fas-associated factor 2) is a ubiquitin ligase adaptor protein that plays a crucial role in the ubiquitin-mediated degradation of misfolded proteins in the endoplasmic reticulum. A recent genome-wide association study indicated an association between FAF2 and ALD; however, the exact contribution of FAF2 to ALD pathogenesis remains unclear. METHODS FAF2 was knocked down using AAV-delivered shRNA in C57/BL6 mice. Mice were subjected to a chronic-plus-single binge ethanol feeding (NIAAA) model. Nine hours after gavage, liver, blood, and other organs of interest were collected for gene expression and biochemical analyses. RESULTS We first observed a significant elevation in hepatic FAF2 protein expression in individuals with ALD and in mice subjected to an ethanol-binge model. Interestingly, knocking down FAF2 in the liver using adeno-associated virus serotype 8-delivered short hairpin RNA conferred a protective effect against alcohol-induced liver steatosis in ethanol-binged mice. Transcriptomic analysis revealed that differentially expressed genes were enriched in multiple lipid metabolism regulation pathways. Further analysis of transcription factors regulating these differentially expressed genes suggested potential regulation by SREBP1. Several SREBP1 target genes, including Fasn, Scd1, Lpin1, and Pcsk9 (proprotein convertase subtilisin/kexin type 9), were dysregulated in the livers of ethanol-fed FAF2 knockdown mice. Additionally, Pcsk9 could be regulated through the FOXO3-SIRT6 pathway in the livers of ethanol-fed FAF2 knockdown mice, leading to increased liver low-density lipoprotein receptor expression and reduced plasma LDL cholesterol levels. Furthermore, FAF2 knockdown in mouse liver enhanced adipose triglyceride lipase lipolytic activity by upregulating the adipose triglyceride lipase activator, comparative gene identification-58, and downregulating the adipose triglyceridelipase transport inhibitor, Elmod2, contributing to the alleviation of liver steatosis. CONCLUSIONS Our study uncovers a novel mechanism involving FAF2 in the pathogenesis of ALD.
Collapse
Affiliation(s)
- Nazmul Huda
- Department of Medicine, Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Praveen Kusumanchi
- Department of Medicine, Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Yanchao Jiang
- Department of Medicine, Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Hui Gao
- Department of Medicine, Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Themis Thoudam
- Department of Medicine, Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Ge Zeng
- Department of Medicine, Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Nicholas J. Skill
- Department of Surgery, Louisiana State University Health Science Center, New Orleans, Louisiana, USA
| | - Zhaoli Sun
- Department of Surgery, John Hopkins University, Baltimore, Maryland, USA
| | - Suthat Liangpunsakul
- Department of Medicine, Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Research, Roudebush Veterans Administration Medical Center, Indianapolis, Indiana, USA
| | - Jing Ma
- Department of Medicine, Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Zhihong Yang
- Department of Medicine, Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
5
|
Xu K, Corona-Avila I, Frutos MD, Nunez-Sanchez MA, Makhanasa D, Shah PV, Guzman G, Ramos-Molina B, Priyadarshini M, Khan MW. Hepatic HKDC1 Deletion Alleviates Western Diet-Induced MASH in Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.11.26.625530. [PMID: 39651120 PMCID: PMC11623584 DOI: 10.1101/2024.11.26.625530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
The global prevalence of Metabolic dysfunction-associated steatohepatitis (MASH) has been rising sharply, closely mirroring the increasing rates of obesity and metabolic syndrome. MASH exhibits a strong sexual dimorphism where females are affected with more severe forms after menopause. Hexokinase domain-containing protein 1 (HKDC1) has recently been recognized for its role in liver diseases, where its expression is minimal under normal conditions but significantly increases in response to metabolic stressors like obesity and liver injury. This selective upregulation suggests HKDC1s potential specialization in hepatic glucose and lipid dysregulation, linking it closely to the progression of MASLD and MASH. This study aims to clarify the role of HKDC1 in Western diet-induced MASH in female mice by examining its impact on hepatic glucose and lipid metabolism, offering insights into its potential as a therapeutic target and addressing the need for sex-specific research in liver disease. This study reveals that HKDC1 expression is elevated in obese women with MASH and correlates with liver pathology. In a mouse model, liver-specific HKDC1 knockout (HKDC1LKO) protected against Western diet-induced obesity, glucose intolerance, and MASH features, including steatosis, inflammation, and fibrosis. Transcriptomic analysis showed that HKDC1 deletion reduced pro-inflammatory and pro-fibrotic gene expression, while gut microbiome analysis indicated a shift toward MASH-protective bacteria. These findings suggest that HKDC1 may exacerbate MASH progression through its role in metabolic and inflammatory pathways, making it a potential therapeutic target.
Collapse
|
6
|
Liu Q, Li P, Ma J, Zhang J, Li W, Liu Y, Liu L, Liang S, He M. Arsenic exposure at environmentally relevant levels induced metabolic toxicity in development mice: Mechanistic insights from integrated transcriptome and metabolome. ENVIRONMENT INTERNATIONAL 2024; 190:108819. [PMID: 38906090 DOI: 10.1016/j.envint.2024.108819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 05/22/2024] [Accepted: 06/11/2024] [Indexed: 06/23/2024]
Abstract
Emerging evidence has linked arsenic exposure and metabolic homeostasis, but the mechanism is incompletely understood, especially at relatively low concentrations. In this study, we used a mouse model to evaluate the health impacts and metabolic toxicity of arsenic exposure in drinking water at environmentally relevant levels (0.25 and 1.0 ppm). Our results indicated that arsenic damaged intestinal barrier and induced arsenic accumulation, oxidative stress, and pathological changes in the liver and illum. Interestingly, arsenic increased the hepatic triglyceride (TG) and total cholesterol (TC), while reduced serum TG and TC levels. The liver transcriptome found that arsenic exposure caused transcriptome perturbation and promoted hepatic lipid accumulation by regulating the exogenous fatty acids degradation and apolipoproteins related genes. The serum metabolomics identified 74 and 88 differential metabolites in 0.25 and 1.0 ppm, respectively. The KEGG disease and subcellular location analysis indicated that arsenic induced liver and intestinal diseases, and the mitochondrion might be the target organelle for arsenic-induced toxicity. Co-enrichment of transcriptome and metabolome identified 24 metabolites and 9 genes as metabolic toxicity biomarkers. Moreover, 40 male (20 nonalcoholic fatty liver disease (NAFLD) cases and 20 healthy controls) was further selected to validate our findings. Importantly, the significantly changed L-palmitoylcarnitine, 3-hydroxybutyric acid, 2-hydroxycaproic acid and 6 genes of Hadha, Acadl, Aldh3a2, Cpt1a, Cpt2, and Acox1 were found in the NAFLD cases. The results from integrated multi-omics and chemical-protein network analysis indicated that L-palmitoylcarnitine played a critical role in metabolic toxicity by regulating mitochondrial fatty acids β-oxidation genes (Cpt1a, Cpt2). In conclusion, these findings provided new clues for the metabolic toxicity of arsenic exposure at environmentally relevant levels, which involved in the late-life NAFLD development. Our results also contribute to understanding the human responses and phenotypic changes to this hazardous material exposure in the environment.
Collapse
Affiliation(s)
- Qianying Liu
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental Health for Incubating, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Peiwen Li
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental Health for Incubating, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jinglan Ma
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental Health for Incubating, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jiazhen Zhang
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental Health for Incubating, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Weiya Li
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental Health for Incubating, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yuenan Liu
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental Health for Incubating, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Lu Liu
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental Health for Incubating, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Sen Liang
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental Health for Incubating, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Meian He
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental Health for Incubating, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
7
|
Zhang Q, Wu S, Chen Q, Zhang Y, Zhang C, Yin R, Ouyang Z, Wei Y. Reducing Oxidative Stress-Mediated Alcoholic Liver Injury by Multiplexed RNAi of Cyp2e1, Cyp4a10, and Cyp4a14. Biomedicines 2024; 12:1505. [PMID: 39062078 PMCID: PMC11274525 DOI: 10.3390/biomedicines12071505] [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: 04/29/2024] [Revised: 06/24/2024] [Accepted: 07/02/2024] [Indexed: 07/28/2024] Open
Abstract
The prevalence of excessive drinking-related alcoholic liver disease (ALD) is rising, yet therapeutic options remain limited. High alcohol consumption and consequent oxidative metabolism by cytochrome P450 (CYP) can lead to extremely high levels of reactive oxygen species, which overwhelm cellular defenses and harm hepatocytes. Our previous investigations showed that inhibiting Cyp2e1 using RNA interference reduced the incidence of ALD. However, compensatory mechanisms other than CYP2E1 contribute to oxidative stress in the liver. Therefore, we coupled triple siRNA lipid nanoparticles (LNPs) targeting Cyp2e1 with two isoenzymes Cyp4a10 and Cyp4a14 to treat ALD mouse models fed with Lieber-Decarli ethanol liquid diet for 12 weeks at the early (1st week), middle (5th week), and late (9th week) stages. The administration of triple siRNA LNPs significantly ameliorated chronic alcoholic liver injury in mice, and early treatment achieved the most profound effects. These effects can be attributed to a reduction in oxidative stress and increased expression of antioxidant genes, including Gsh-Px, Gsh-Rd, and Sod1. Moreover, we observed the alleviation of inflammation, evidenced by the downregulation of Il-1β, Il-6, Tnf-α, and Tgf-β, and the prevention of excessive lipid synthesis, evidenced by the restoration of the expression of Srebp1c, Acc, and Fas. Finally, triple siRNA treatment maintained normal metabolism in lipid oxidation. In brief, our research examined the possible targets for clinical intervention in ALD by examining the therapeutic effects of triple siRNA LNPs targeting Cyp2e1, Cyp4a10, and Cyp4a14. The in vivo knockdown of the three genes in this study is suggested as a promising siRNA therapeutic approach for ALD.
Collapse
Affiliation(s)
- Qi Zhang
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, China; (Q.Z.); (S.W.)
| | - Shuang Wu
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, China; (Q.Z.); (S.W.)
| | - Qiubing Chen
- Department of Urology, Frontier Science Centre for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Yahong Zhang
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, China; (Q.Z.); (S.W.)
| | - Cai Zhang
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, China; (Q.Z.); (S.W.)
| | - Runting Yin
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, China; (Q.Z.); (S.W.)
| | - Zhen Ouyang
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, China; (Q.Z.); (S.W.)
| | - Yuan Wei
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, China; (Q.Z.); (S.W.)
| |
Collapse
|
8
|
Hu J, Li S, Zhong X, Wei Y, Sun Q, Zhong L. Human umbilical cord mesenchymal stem cells attenuate diet-induced obesity and NASH-related fibrosis in mice. Heliyon 2024; 10:e25460. [PMID: 38356602 PMCID: PMC10864966 DOI: 10.1016/j.heliyon.2024.e25460] [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: 09/12/2023] [Revised: 01/26/2024] [Accepted: 01/26/2024] [Indexed: 02/16/2024] Open
Abstract
Non-alcoholic steatohepatitis (NASH) is a progressive form of non-alcoholic fatty liver disease (NAFLD) that may progress to cirrhosis and hepatocellular carcinoma but has no available treatment. Mesenchymal stem cells (MSCs) have become increasingly prominent in cell therapy. Human umbilical cord MSCs (hUC-MSCs) are considered superior to other MSCs due to their strong immunomodulatory ability, ease of collection, low immune rejection, and no tumorigenicity. Though hUC-MSCs have received increasing attention in research, they have been rarely applied in any investigations or treatments of NASH and associated fibrosis. Therefore, this study evaluated the therapeutic efficacy of hUC-MSCs in C57BL/6 mice with diet-induced NASH. At week 32, mice were randomized into two groups: phosphate-buffered saline and MSCs, which were injected into the tail vein. At week 40, glucose metabolism was evaluated using glucose and insulin tolerance tests. NASH-related indicators were examined using various biological methods. hUC-MSC administration alleviated obesity, glucose metabolism, hepatic steatosis, inflammation, and fibrosis. Liver RNA-seq showed that the expression of the acyl-CoA thioesterase (ACOT) family members Acot1, Acot2, and Acot3 involved in fatty acid metabolism were altered. The cytochrome P450 (CYP) members Cyp4a10 and Cyp4a14, which are involved in the peroxisome proliferator-activator receptor (PPAR) signaling pathway, were significantly downregulated after hUC-MSC treatment. In conclusion, hUC-MSCs effectively reduced Western diet-induced obesity, NASH, and fibrosis in mice, partly by regulating lipid metabolism and the PPAR signaling pathway.
Collapse
Affiliation(s)
- Jiali Hu
- Department of Gastroenterology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shan Li
- Department of Gastroenterology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xuan Zhong
- Department of Gastroenterology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yushuang Wei
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, 200120, China
| | - Qinjuan Sun
- Department of Gastroenterology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lan Zhong
- Department of Gastroenterology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, 200120, China
| |
Collapse
|
9
|
Abstract
Alcohol-associated liver disease (ALD) is a major cause of chronic liver disease worldwide, and comprises a spectrum of several different disorders, including simple steatosis, steatohepatitis, cirrhosis, and superimposed hepatocellular carcinoma. Although tremendous progress has been made in the field of ALD over the last 20 years, the pathogenesis of ALD remains obscure, and there are currently no FDA-approved drugs for the treatment of ALD. In this Review, we discuss new insights into the pathogenesis and therapeutic targets of ALD, utilizing the study of multiomics and other cutting-edge approaches. The potential translation of these studies into clinical practice and therapy is deliberated. We also discuss preclinical models of ALD, interplay of ALD and metabolic dysfunction, alcohol-associated liver cancer, the heterogeneity of ALD, and some potential translational research prospects for ALD.
Collapse
|
10
|
Zhang-Sun ZY, Xu XZ, Escames G, Lei WR, Zhao L, Zhou YZ, Tian Y, Ren YN, Acuña-Castroviejo D, Yang Y. Targeting NR1D1 in organ injury: challenges and prospects. Mil Med Res 2023; 10:62. [PMID: 38072952 PMCID: PMC10712084 DOI: 10.1186/s40779-023-00495-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 11/13/2023] [Indexed: 12/18/2023] Open
Abstract
Nuclear receptor subfamily 1, group D, member 1 (NR1D1, also known as REV-ERBα) belongs to the nuclear receptor (NR) family, and is a heme-binding component of the circadian clock that consolidates circadian oscillators. In addition to repressing the transcription of multiple clock genes associated with circadian rhythms, NR1D1 has a wide range of downstream target genes that are intimately involved in many physiopathological processes, including autophagy, immunity, inflammation, metabolism and aging in multiple organs. This review focuses on the pivotal role of NR1D1 as a key transcription factor in the gene regulatory network, with particular emphasis on the milestones of the latest discoveries of NR1D1 ligands. NR1D1 is considered as a promising drug target for treating diverse diseases and may contribute to research on innovative biomarkers and therapeutic targets for organ injury-related diseases. Further research on NR1D1 ligands in prospective human trials may pave the way for their clinical application in many organ injury-related disorders.
Collapse
Affiliation(s)
- Zi-Yin Zhang-Sun
- Department of Cardiology, Northwest University First Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine , Northwest University, Xi'an, 710069, China
| | - Xue-Zeng Xu
- Department of Cardiovascular Surgery, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Germaine Escames
- Biomedical Research Center, Department of Physiology, Faculty of Medicine, Institute of Biotechnology, Technological Park of Health Sciences, University of Granada, 18016, Granada, Spain
- Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Ibs.Granada, San Cecilio University Hospital, 18016, Granada, Spain
| | - Wang-Rui Lei
- Department of Cardiology, Northwest University First Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine , Northwest University, Xi'an, 710069, China
| | - Lin Zhao
- Department of Cardiovascular Surgery, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Ya-Zhe Zhou
- Department of Cardiology, Northwest University First Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine , Northwest University, Xi'an, 710069, China
| | - Ye Tian
- Department of Cardiology, Northwest University First Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine , Northwest University, Xi'an, 710069, China
| | - Ya-Nan Ren
- Department of Cardiology, Northwest University First Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine , Northwest University, Xi'an, 710069, China
| | - Darío Acuña-Castroviejo
- Biomedical Research Center, Department of Physiology, Faculty of Medicine, Institute of Biotechnology, Technological Park of Health Sciences, University of Granada, 18016, Granada, Spain.
- Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Ibs.Granada, San Cecilio University Hospital, 18016, Granada, Spain.
- UGC of Clinical Laboratories, San Cecilio Clinical University Hospital, 18016, Granada, Spain.
| | - Yang Yang
- Department of Cardiology, Northwest University First Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China.
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine , Northwest University, Xi'an, 710069, China.
| |
Collapse
|
11
|
Zhong D, Cai J, Hu C, Chen J, Zhang R, Fan C, Li S, Zhang H, Xu Z, Jia Z, Guo D, Sun Y. Inhibition of mPGES-2 ameliorates NASH by activating NR1D1 via heme. Hepatology 2023; 78:547-561. [PMID: 35839302 DOI: 10.1002/hep.32671] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 06/27/2022] [Accepted: 07/12/2022] [Indexed: 12/08/2022]
Abstract
BACKGROUND AND AIMS Nonalcoholic fatty liver disease (NAFLD), a complex metabolic syndrome, has limited therapeutic options. Microsomal prostaglandin E synthase-2 (mPGES-2) was originally discovered as a prostaglandin E 2 (PGE 2 ) synthase; however, it does not produce PGE 2 in the liver. Moreover, the role of mPGES-2 in NAFLD remains undefined. Herein, we aimed to determine the function and mechanism of mPGES-2 in liver steatosis and steatohepatitis. APPROACH AND RESULTS To evaluate the role of mPGES-2 in NAFLD, whole-body or hepatocyte-specific mPGES-2-deficient mice fed a high-fat or methionine-choline-deficient diet were used. Compared with control mice, mPGES-2-deficient mice showed reduced hepatic lipid accumulation, along with ameliorated liver injury, inflammation, and fibrosis. Furthermore, the protective effect of mPGES-2 deficiency against NAFLD was dependent on decreased cytochrome P450 4A14 and increased acyl-CoA thioesterase 4 levels regulated by the heme receptor nuclear receptor subfamily 1 group D member 1 (NR1D1), but not PGE 2 . Heme regulated the increased NR1D1 activity mediated by mPGES-2 deficiency. Further, we confirmed the protective role of the mPGES-2 inhibitor SZ0232 in NAFLD therapy. CONCLUSION Our study indicates the pathogenic role of mPGES-2 and outlines the mechanism in mediating NAFLD, thereby highlighting the therapeutic potential of mPGES-2 inhibition in liver steatosis and steatohepatitis.
Collapse
Affiliation(s)
- Dandan Zhong
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy , Xuzhou Medical University , Xuzhou , Jiangsu , P. R. China
| | - Jie Cai
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy , Xuzhou Medical University , Xuzhou , Jiangsu , P. R. China
- Nanjing Key Laboratory of Pediatrics , Nanjing Children's Hospital , Nanjing Medical University , Nanjing , Jiangsu , P. R. China
- Public Experimental Research Center of Xuzhou Medical University , Xuzhou Medical University , Xuzhou , Jiangsu , P. R. China
| | - Cheng Hu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy , Xuzhou Medical University , Xuzhou , Jiangsu , P. R. China
| | - Jingshuo Chen
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy , Xuzhou Medical University , Xuzhou , Jiangsu , P. R. China
- Public Experimental Research Center of Xuzhou Medical University , Xuzhou Medical University , Xuzhou , Jiangsu , P. R. China
| | - Rumeng Zhang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy , Xuzhou Medical University , Xuzhou , Jiangsu , P. R. China
- Department of Pharmacology , Xuzhou Central Hospital , Xuzhou , Jiangsu , China
| | - Chenyu Fan
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy , Xuzhou Medical University , Xuzhou , Jiangsu , P. R. China
| | - Shanshan Li
- Jiangsu Medical Engineering Research Center of Gene Detection , Xuzhou , Jiangsu , China
| | - Hongxing Zhang
- Jiangsu Province Key Laboratory of Anesthesiology , Xuzhou Medical University , Xuzhou , Jiangsu , P. R. China
| | - Zhou Xu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy , Xuzhou Medical University , Xuzhou , Jiangsu , P. R. China
- Public Experimental Research Center of Xuzhou Medical University , Xuzhou Medical University , Xuzhou , Jiangsu , P. R. China
| | - Zhanjun Jia
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy , Xuzhou Medical University , Xuzhou , Jiangsu , P. R. China
- Nanjing Key Laboratory of Pediatrics , Nanjing Children's Hospital , Nanjing Medical University , Nanjing , Jiangsu , P. R. China
| | - Dong Guo
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy , Xuzhou Medical University , Xuzhou , Jiangsu , P. R. China
| | - Ying Sun
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy , Xuzhou Medical University , Xuzhou , Jiangsu , P. R. China
| |
Collapse
|
12
|
Li X, Zheng T, Zhang Y, Zhao Y, Liu F, Dai S, Liu X, Zhang M. Dickkopf-1 promotes vascular smooth muscle cell foam cell formation and atherosclerosis development through CYP4A11/SREBP2/ABCA1. FASEB J 2023; 37:e23048. [PMID: 37389895 DOI: 10.1096/fj.202300295r] [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: 02/18/2023] [Revised: 05/20/2023] [Accepted: 06/08/2023] [Indexed: 07/01/2023]
Abstract
Vascular smooth muscle cells (VSMCs) are considered to be a crucial source of foam cells in atherosclerosis due to their low expression level of cholesterol exporter ATP-binding cassette transporter A1 (ABCA1) intrinsically. While the definite regulatory mechanisms are complicated and have not yet been fully elucidated, we previously reported that Dickkopf-1 (DKK1) mediates endothelial cell (EC) dysfunction, thereby aggravating atherosclerosis. However, the role of smooth muscle cell (SMC) DKK1 in atherosclerosis and foam cell formation remains unknown. In this study, we established SMC-specific DKK1-knockout (DKK1SMKO ) mice by crossbreeding DKK1flox/flox mice with TAGLN-Cre mice. Then, DKK1SMKO mice were crossed with APOE-/- mice to generate DKK1SMKO /APOE-/- mice, which exhibited milder atherosclerotic burden and fewer SMC foam cells. In vitro loss- and gain-of-function studies of DKK1 in primary human aortic smooth muscle cells (HASMCs) have proven that DKK1 prevented oxidized lipid-induced ABCA1 upregulation and cholesterol efflux and promoted SMC foam cell formation. Mechanistically, RNA-sequencing (RNA-seq) analysis of HASMCs as well as chromatin immunoprecipitation (ChIP) experiments showed that DKK1 mediates the binding of transcription factor CCAAT/enhancer-binding protein delta (C/EBPδ) to the promoter of cytochrome P450 epoxygenase 4A11 (CYP4A11) to regulate its expression. In addition, CYP4A11 as well as its metabolite 20-HETE-promoted activation of transcription factor sterol regulatory element-binding protein 2 (SREBP2) mediated the DKK1 regulation of ABCA1 in SMC. Furthermore, HET0016, the antagonist of CYP4A11, has also shown an alleviating effect on atherosclerosis. In conclusion, our results demonstrate that DKK1 promotes SMC foam cell formation during atherosclerosis via a reduction in CYP4A11-20-HETE/SREBP2-mediated ABCA1 expression.
Collapse
Affiliation(s)
- Xiao Li
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Tengfei Zheng
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Yu Zhang
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Yachao Zhao
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Fengming Liu
- Department of Immunology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Shen Dai
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Xiaolin Liu
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Mei Zhang
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
13
|
Ni Y, Nan S, Zheng L, Zhang L, Zhao Y, Fu Z. Time-dependent effect of REV-ERBα agonist SR9009 on nonalcoholic steatohepatitis and gut microbiota in mice. Chronobiol Int 2023; 40:769-782. [PMID: 37161366 DOI: 10.1080/07420528.2023.2207649] [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: 01/06/2023] [Revised: 04/17/2023] [Accepted: 04/21/2023] [Indexed: 05/11/2023]
Abstract
The circadian clock is involved in the pathogenesis of nonalcoholic steatohepatitis (NASH), and the target pathways of many NASH candidate drugs are controlled by the circadian clock. However, the application of chronopharmacology in NASH is little considered currently. Here, the time-dependent effect of REV-ERBα agonist SR9009 on diet-induced NASH and microbiota was investigated. C57BL/6J mice were fed a high-cholesterol and high-fat diet (CL) for 12 weeks to induce NASH and then treated with SR9009 either at Zeitgeber time 0 (ZT0) or ZT12 for another 6 weeks. Pharmacological activation of REV-ERBα by SR9009 alleviated hepatic steatosis, insulin resistance, liver inflammation, and fibrosis in CL diet-induced NASH mice. These effects were accompanied by improved gut barrier function and altered microbial composition and function in NASH mice, and the effect tended to be stronger when SR9009 was injected at ZT0. Moreover, SR9009 treatment at different time points resulted in a marked difference in the composition of the microbiota, with a stronger effect on the enrichment of beneficial bacteria and the diminishment of harmful bacteria when SR9009 was administrated at ZT0. Therefore, the time-dependent effect of REV-ERBα agonist on NASH was partly associated with the microbiota, highlighting the potential role of microbiota in the chronopharmacology of NASH and the possibility of discovering new therapeutic strategies for NASH.
Collapse
Affiliation(s)
- Yinhua Ni
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
| | - Sujie Nan
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
| | - Liujie Zheng
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
| | - Liqian Zhang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
| | - Yufeng Zhao
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
| | - Zhengwei Fu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
| |
Collapse
|
14
|
Cao L, Wu D, Qin L, Tan D, Fan Q, Jia X, Yang M, Zhou T, Feng C, Lu Y, He Y. Single-Cell RNA Transcriptome Profiling of Liver Cells of Short-Term Alcoholic Liver Injury in Mice. Int J Mol Sci 2023; 24:ijms24054344. [PMID: 36901774 PMCID: PMC10002329 DOI: 10.3390/ijms24054344] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 01/23/2023] [Accepted: 02/20/2023] [Indexed: 02/24/2023] Open
Abstract
Alcoholic liver disease (ALD) is currently considered a global healthcare problem with limited pharmacological treatment options. There are abundant cell types in the liver, such as hepatocytes, endothelial cells, Kupffer cells and so on, but little is known about which kind of liver cells play the most important role in the process of ALD. To obtain a cellular resolution of alcoholic liver injury pathogenesis, 51,619 liver single-cell transcriptomes (scRNA-seq) with different alcohol consumption durations were investigated, 12 liver cell types were identified, and the cellular and molecular mechanisms of the alcoholic liver injury were revealed. We found that more aberrantly differential expressed genes (DEGs) were present in hepatocytes, endothelial cells, and Kupffer cells than in other cell types in alcoholic treatment mice. Alcohol promoted the pathological processes of liver injury; the specific mechanisms involved: lipid metabolism, oxidative stress, hypoxia, complementation and anticoagulation, and hepatocyte energy metabolism on hepatocytes; NO production, immune regulation, epithelial and cell migration on endothelial cells; antigen presentation and energy metabolism on Kupffer cells, based on the GO analysis. In addition, our results showed that some transcription factors (TFs) are activated in alcohol-treated mice. In conclusion, our study improves the understanding of liver cell heterogeneity in alcohol-fed mice at the single-cell level. It has potential value for understanding key molecular mechanisms and improving current prevention and treatment strategies for short-term alcoholic liver injury.
Collapse
Affiliation(s)
- Ligang Cao
- Guizhou Engineering Research Center of Industrial Key-Technology for Dendrobium Nobile, Zunyi Medical University, Zunyi 563000, China
| | - Di Wu
- Guizhou Engineering Research Center of Industrial Key-Technology for Dendrobium Nobile, Zunyi Medical University, Zunyi 563000, China
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563000, China
| | - Lin Qin
- Guizhou Engineering Research Center of Industrial Key-Technology for Dendrobium Nobile, Zunyi Medical University, Zunyi 563000, China
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563000, China
| | - Daopeng Tan
- Guizhou Engineering Research Center of Industrial Key-Technology for Dendrobium Nobile, Zunyi Medical University, Zunyi 563000, China
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563000, China
| | - Qingjie Fan
- Guizhou Engineering Research Center of Industrial Key-Technology for Dendrobium Nobile, Zunyi Medical University, Zunyi 563000, China
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563000, China
| | - Xiaohuan Jia
- Guizhou Engineering Research Center of Industrial Key-Technology for Dendrobium Nobile, Zunyi Medical University, Zunyi 563000, China
| | - Mengting Yang
- Guizhou Engineering Research Center of Industrial Key-Technology for Dendrobium Nobile, Zunyi Medical University, Zunyi 563000, China
| | - Tingting Zhou
- Guizhou Engineering Research Center of Industrial Key-Technology for Dendrobium Nobile, Zunyi Medical University, Zunyi 563000, China
| | - Chengcheng Feng
- Guizhou Engineering Research Center of Industrial Key-Technology for Dendrobium Nobile, Zunyi Medical University, Zunyi 563000, China
| | - Yanliu Lu
- Guizhou Engineering Research Center of Industrial Key-Technology for Dendrobium Nobile, Zunyi Medical University, Zunyi 563000, China
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563000, China
| | - Yuqi He
- Guizhou Engineering Research Center of Industrial Key-Technology for Dendrobium Nobile, Zunyi Medical University, Zunyi 563000, China
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563000, China
- Correspondence:
| |
Collapse
|
15
|
Zhang H, Zhang J, Li J, Mao Z, Qian J, Zong C, Sun H, Yuan B. Multi-Omics Analyses Reveal the Mechanisms of Early Stage Kidney Toxicity by Diquat. TOXICS 2023; 11:184. [PMID: 36851058 PMCID: PMC9966843 DOI: 10.3390/toxics11020184] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/11/2023] [Accepted: 02/14/2023] [Indexed: 06/18/2023]
Abstract
Diquat (DQ), a widely used bipyridyl herbicide, is associated with significantly higher rates of kidney injuries compared to other pesticides. However, the underlying molecular mechanisms are largely unknown. In this study, we identified the molecular changes in the early stage of DQ-induced kidney damage in a mouse model through transcriptomic, proteomic and metabolomic analyses. We identified 869 genes, 351 proteins and 96 metabolites that were differentially expressed in the DQ-treated mice relative to the control mice (p < 0.05), and showed significant enrichment in the PPAR signaling pathway and fatty acid metabolism. Hmgcs2, Cyp4a10, Cyp4a14 and Lpl were identified as the major proteins/genes associated with DQ-induced kidney damage. In addition, eicosapentaenoic acid, linoleic acid, palmitic acid and (R)-3-hydroxybutyric acid were the major metabolites related to DQ-induced kidney injury. Overall, the multi-omics analysis showed that DQ-induced kidney damage is associated with dysregulation of the PPAR signaling pathway, and an aberrant increase in Hmgcs2 expression and 3-hydroxybutyric acid levels. Our findings provide new insights into the molecular basis of DQ-induced early kidney damage.
Collapse
Affiliation(s)
- Huazhong Zhang
- Department of Emergency, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- Institute of Poisoning, Nanjing Medical University, Nanjing 211100, China
| | - Jinsong Zhang
- Department of Emergency, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- Institute of Poisoning, Nanjing Medical University, Nanjing 211100, China
| | - Jinquan Li
- Department of Emergency, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- Institute of Poisoning, Nanjing Medical University, Nanjing 211100, China
| | - Zhengsheng Mao
- Institute of Poisoning, Nanjing Medical University, Nanjing 211100, China
| | - Jian Qian
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Cheng Zong
- College of Safety Science and Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Hao Sun
- Department of Emergency, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- Institute of Poisoning, Nanjing Medical University, Nanjing 211100, China
| | - Beilei Yuan
- College of Safety Science and Engineering, Nanjing Tech University, Nanjing 211816, China
| |
Collapse
|
16
|
Kyritsi K, Wu N, Zhou T, Carpino G, Baiocchi L, Kennedy L, Chen L, Ceci L, Meyer AA, Barupala N, Franchitto A, Onori P, Ekser B, Gaudio E, Wu C, Marakovits C, Chakraborty S, Francis H, Glaser S, Alpini G. Knockout of secretin ameliorates biliary and liver phenotypes during alcohol-induced hepatotoxicity. Cell Biosci 2023; 13:5. [PMID: 36624475 PMCID: PMC9830859 DOI: 10.1186/s13578-022-00945-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 12/19/2022] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Alcohol-related liver disease (ALD) is characterized by ductular reaction (DR), liver inflammation, steatosis, fibrosis, and cirrhosis. The secretin (Sct)/secretin receptor (SR) axis (expressed only by cholangiocytes) regulates liver phenotypes in cholestasis. We evaluated the role of Sct signaling on ALD phenotypes. METHODS We used male wild-type and Sct-/- mice fed a control diet (CD) or ethanol (EtOH) for 8 wk. Changes in liver phenotypes were measured in mice, female/male healthy controls, and patients with alcoholic cirrhosis. Since Cyp4a10 and Cyp4a11/22 regulate EtOH liver metabolism, we measured their expression in mouse/human liver. We evaluated: (i) the immunoreactivity of the lipogenesis enzyme elongation of very-long-chain fatty acids 1 (Elovl, mainly expressed by hepatocytes) in mouse/human liver sections by immunostaining; (ii) the expression of miR-125b (that is downregulated in cholestasis by Sct) in mouse liver by qPCR; and (iii) total bile acid (BA) levels in mouse liver by enzymatic assay, and the mRNA expression of genes regulating BA synthesis (cholesterol 7a-hydroxylase, Cyp27a1, 12a-hydroxylase, Cyp8b1, and oxysterol 7a-hydroxylase, Cyp7b11) and transport (bile salt export pump, Bsep, Na+-taurocholate cotransporting polypeptide, NTCP, and the organic solute transporter alpha (OSTa) in mouse liver by qPCR. RESULTS In EtOH-fed WT mice there was increased biliary and liver damage compared to control mice, but decreased miR-125b expression, phenotypes that were blunted in EtOH-fed Sct-/- mice. The expression of Cyp4a10 increased in cholangiocytes and hepatocytes from EtOH-fed WT compared to control mice but decreased in EtOH-fed Sct-/- mice. There was increased immunoreactivity of Cyp4a11/22 in patients with alcoholic cirrhosis compared to controls. The expression of miR-125b decreased in EtOH-fed WT mice but returned at normal values in EtOH-fed Sct-/- mice. Elovl1 immunoreactivity increased in patients with alcoholic cirrhosis compared to controls. There was no difference in BA levels between WT mice fed CD or EtOH; BA levels decreased in EtOH-fed Sct-/- compared to EtOH-fed WT mice. There was increased expression of Cyp27a1, Cyp8b1, Cyp7b1, Bsep, NTCP and Osta in total liver from EtOH-fed WT compared to control mice, which decreased in EtOH-fed Sct-/- compared to EtOH-fed WT mice. CONCLUSIONS Targeting Sct/SR signaling may be important for modulating ALD phenotypes.
Collapse
Affiliation(s)
- Konstantina Kyritsi
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Nan Wu
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Tianhao Zhou
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Guido Carpino
- Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences, La Sapienza University of Rome, Rome, Italy
| | | | - Lindsey Kennedy
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
- Division of Research, Indiana Center for Liver Research, Gastroenterology, Medicine, Richard L. Roudebush VA Medical Center and Indiana University, 702 Rotary Circle, Rm. 013C, Indianapolis, IN, 46202-2859, USA
| | - Lixian Chen
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ludovica Ceci
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences, La Sapienza University of Rome, Rome, Italy
| | - Alison Ann Meyer
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Nipuni Barupala
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Antonio Franchitto
- Department of Movement, Human and Health Sciences, University of Rome "Foro Italico", Rome, Italy
| | - Paolo Onori
- Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences, La Sapienza University of Rome, Rome, Italy
| | - Burcin Ekser
- Division of Transplant Surgery, Department of Surgery, Indiana University, Indianapolis, IN, USA
| | - Eugenio Gaudio
- Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences, La Sapienza University of Rome, Rome, Italy
| | - Chaodong Wu
- Department of Nutrition, Texas A&M University, College Station, TX, USA
| | - Corinn Marakovits
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sanjukta Chakraborty
- Department of Medical Physiology, Texas A&M University School of Medicine, 8447 Riverside Parkway, MREB II, Room 2342, Bryan, TX, 77807-3260, USA
| | - Heather Francis
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA.
- Division of Research, Indiana Center for Liver Research, Gastroenterology, Medicine, Richard L. Roudebush VA Medical Center and Indiana University, 702 Rotary Circle, Rm. 013C, Indianapolis, IN, 46202-2859, USA.
| | - Shannon Glaser
- Department of Medical Physiology, Texas A&M University School of Medicine, 8447 Riverside Parkway, MREB II, Room 2342, Bryan, TX, 77807-3260, USA.
| | - Gianfranco Alpini
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA.
- Division of Research, Indiana Center for Liver Research, Gastroenterology, Medicine, Richard L. Roudebush VA Medical Center and Indiana University, 702 Rotary Circle, Rm. 013C, Indianapolis, IN, 46202-2859, USA.
| |
Collapse
|
17
|
ZHOU T, CAO L, QIN L, LU Y, HE Y, TAN D. Differential effects of medium- and long-term high-fat diets on the expression of genes or proteins related to nonalcoholic fatty liver disease in mice. FOOD SCIENCE AND TECHNOLOGY 2023. [DOI: 10.1590/fst.117522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
|
18
|
Murray MH, Valfort AC, Koelblen T, Ronin C, Ciesielski F, Chatterjee A, Veerakanellore GB, Elgendy B, Walker JK, Hegazy L, Burris TP. Structural basis of synthetic agonist activation of the nuclear receptor REV-ERB. Nat Commun 2022; 13:7131. [PMID: 36414641 PMCID: PMC9681850 DOI: 10.1038/s41467-022-34892-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 11/10/2022] [Indexed: 11/23/2022] Open
Abstract
The nuclear receptor REV-ERB plays an important role in a range of physiological processes. REV-ERB behaves as a ligand-dependent transcriptional repressor and heme has been identified as a physiological agonist. Our current understanding of how ligands bind to and regulate transcriptional repression by REV-ERB is based on the structure of heme bound to REV-ERB. However, porphyrin (heme) analogues have been avoided as a source of synthetic agonists due to the wide range of heme binding proteins and potential pleotropic effects. How non-porphyrin synthetic agonists bind to and regulate REV-ERB has not yet been defined. Here, we characterize a high affinity synthetic REV-ERB agonist, STL1267, and describe its mechanism of binding to REV-ERB as well as the method by which it recruits transcriptional corepressor both of which are unique and distinct from that of heme-bound REV-ERB.
Collapse
Affiliation(s)
- Meghan H Murray
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO, 63104, USA
- Center for Clinical Pharmacology, Washington University School of Medicine, University of Health Sciences & Pharmacy, St. Louis, MO, 63110, USA
| | | | - Thomas Koelblen
- University of Florida Genetics Institute, Gainesville, FL, 32610, USA
| | | | | | - Arindam Chatterjee
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO, 63104, USA
| | - Giri Babu Veerakanellore
- Center for Clinical Pharmacology, Washington University School of Medicine, University of Health Sciences & Pharmacy, St. Louis, MO, 63110, USA
- Department of Pharmaceutical and Administrative Sciences, University of Health Sciences & Pharmacy, St. Louis, MO, 63110, USA
| | - Bahaa Elgendy
- Center for Clinical Pharmacology, Washington University School of Medicine, University of Health Sciences & Pharmacy, St. Louis, MO, 63110, USA
- Department of Pharmaceutical and Administrative Sciences, University of Health Sciences & Pharmacy, St. Louis, MO, 63110, USA
| | - John K Walker
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO, 63104, USA
| | - Lamees Hegazy
- Center for Clinical Pharmacology, Washington University School of Medicine, University of Health Sciences & Pharmacy, St. Louis, MO, 63110, USA.
- Department of Pharmaceutical and Administrative Sciences, University of Health Sciences & Pharmacy, St. Louis, MO, 63110, USA.
| | - Thomas P Burris
- University of Florida Genetics Institute, Gainesville, FL, 32610, USA.
| |
Collapse
|
19
|
Raza GS, Sodum N, Kaya Y, Herzig KH. Role of Circadian Transcription Factor Rev-Erb in Metabolism and Tissue Fibrosis. Int J Mol Sci 2022; 23:12954. [PMID: 36361737 PMCID: PMC9655416 DOI: 10.3390/ijms232112954] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/21/2022] [Accepted: 10/22/2022] [Indexed: 09/12/2023] Open
Abstract
Circadian rhythms significantly affect metabolism, and their disruption leads to cardiometabolic diseases and fibrosis. The clock repressor Rev-Erb is mainly expressed in the liver, heart, lung, adipose tissue, skeletal muscles, and brain, recognized as a master regulator of metabolism, mitochondrial biogenesis, inflammatory response, and fibrosis. Fibrosis is the response of the body to injuries and chronic inflammation with the accumulation of extracellular matrix in tissues. Activation of myofibroblasts is a key factor in the development of organ fibrosis, initiated by hormones, growth factors, inflammatory cytokines, and mechanical stress. This review summarizes the importance of Rev-Erb in ECM remodeling and tissue fibrosis. In the heart, Rev-Erb activation has been shown to alleviate hypertrophy and increase exercise capacity. In the lung, Rev-Erb agonist reduced pulmonary fibrosis by suppressing fibroblast differentiation. In the liver, Rev-Erb inhibited inflammation and fibrosis by diminishing NF-κB activity. In adipose tissue, Rev- Erb agonists reduced fat mass. In summary, the results of multiple studies in preclinical models demonstrate that Rev-Erb is an attractive target for positively influencing dysregulated metabolism, inflammation, and fibrosis, but more specific tools and studies would be needed to increase the information base for the therapeutic potential of these substances interfering with the molecular clock.
Collapse
Affiliation(s)
- Ghulam Shere Raza
- Research Unit of Biomedicine, Medical Research Center, Faculty of Medicine, University of Oulu, 90220 Oulu, Finland
| | - Nalini Sodum
- Research Unit of Biomedicine, Medical Research Center, Faculty of Medicine, University of Oulu, 90220 Oulu, Finland
| | - Yagmur Kaya
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Marmara University, 34854 Istanbul, Turkey
| | - Karl-Heinz Herzig
- Research Unit of Biomedicine, Medical Research Center, Faculty of Medicine, University of Oulu, 90220 Oulu, Finland
- Oulu University Hospital, University of Oulu, 90220 Oulu, Finland
- Pediatric Gastroenterology and Metabolic Diseases, Pediatric Institute, Poznan University of Medical Sciences, 60-572 Poznań, Poland
| |
Collapse
|
20
|
RNA-Seq Analysis of Protection against Chronic Alcohol Liver Injury by Rosa roxburghii Fruit Juice (Cili) in Mice. Nutrients 2022; 14:nu14091974. [PMID: 35565941 PMCID: PMC9104053 DOI: 10.3390/nu14091974] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 05/06/2022] [Accepted: 05/06/2022] [Indexed: 01/27/2023] Open
Abstract
Rosa roxburghii Tratt. fruit juice (Cili) is used as a medicinal and edible resource in China due to its antioxidant and hypolipidemic potentials. The efficacy of Cili in protecting alcohol-induced liver injury and its underlying mechanism was investigated. C57BL/6J mice received a Lieber-DeCarli liquid diet containing alcohol to produce liver injury. After the mice were adapted gradually to 5% alcohol, Cili (4 mL and 8 mL/kg/day for 4 weeks) were gavaged for treatment. The serum enzyme activities, triglyceride levels, histopathology and Oil-red O staining were examined. The RNA-Seq and qPCR analyses were performed to determine the protection mechanisms. Cili decreased serum and liver triglyceride levels in mice receiving alcohol. Hepatocyte degeneration and steatosis were improved by Cili. The RNA-Seq analyses showed Cili brought the alcohol-induced aberrant gene pattern towards normal. The qPCR analysis verified that over-activation of CAR and PXR (Cyp2a4, Cyp2b10 and Abcc4) was attenuated by Cili. Cili alleviated overexpression of oxidative stress responsive genes (Hmox1, Gsta1, Gstm3, Nqo1, Gclc, Vldlr, and Cdkn1a), and rescued alcohol-downregulated metabolism genes (Angptl8, Slc10a2, Ces3b, Serpina12, C6, and Selenbp2). Overall, Cili was effective against chronic alcohol liver injury, and the mechanisms were associated with decreased oxidative stress, improved lipid metabolism through modulating nuclear receptor CAR-, PXR-and Nrf2-mediated pathways.
Collapse
|