1
|
Chen Z, Yan D, Guo S, Song Y, Zhang X, Gu W, Dong H, Huang L. METTL3/miR-192-5p/SCD1 Axis Regulates Lipid Metabolism to Affect T Cell Differentiation in Asthma. Mediators Inflamm 2025; 2025:4955849. [PMID: 39867638 PMCID: PMC11769594 DOI: 10.1155/mi/4955849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 12/10/2024] [Indexed: 01/28/2025] Open
Abstract
Background: This study aimed to explore the mechanisms underlying T-cell differentiation in asthma. Methods and Results: Flow cytometry was performed to detect Th cells. LC-MS/MS was performed to assess lipid metabolism. HE staining was performed to assess the pathological changes of the lung tissues. ELISA was performed to detect cytokine levels. The results of quantitative real-time polymerase chain reaction (qRT-PCR) and western blot showed that miR-192-5p expression was decreased, while SCD1 expression was increased in CD4+T cells isolated from the peripheral blood of children with asthma. The dual luciferase reporter assay determined the direct interaction between miR-192-5p and SCD1. MiR-192-5p inhibitor reduced ASCL3 and PPARα, increased FASN and SREBP1c mRNA expression and protein levels in mouse spleen CD4+T cells, and elevated Th2 and Th17 cells, but these effects were reversed by the SCD1 inhibitor. Oleic acid (OA) reduced Th1 cells and increased Th2 and Th17 cells in mouse spleen CD4+T cells treated with an SCD1 inhibitor. Additionally, pri-miR-192-5p expression was increased in CD4+T cells isolated from the peripheral blood of asthmatic children, and the deletion of METTL3 upregulated pri-miR-192-5p expression in an m6A-dependent manner. MiR-192-5p mimic and inhibitor both reversed miR-192-5p and SCD1 expression affected by overexpression or deletion of METTL3, both in vivo and in vitro. Furthermore, METTL3 overexpression attenuated lung inflammation, elevated Th1 cells, and reduced Th2 and Th17 cells in CD4+T cells isolated from the peripheral blood of asthmatic mice. These effects were reversed by the miR-192-5p inhibitor. Conclusion: These results suggest that METTL3/miR-192-5p/SCD1 axis regulates lipid metabolism and affects T cell differentiation, thus affecting asthma progression. This study may provide novel insights into the pathogenesis of asthma and a new treatment strategy.
Collapse
Affiliation(s)
- Zhengrong Chen
- Department of Pediatric Pulmonology, Children's Hospital of Soochow University, No 303, Jingde Road, Suzhou 215003, China
| | - Dingwei Yan
- Department of Pediatric Pulmonology, Children's Hospital of Soochow University, No 303, Jingde Road, Suzhou 215003, China
| | - Suyu Guo
- Department of Pediatric Pulmonology, Xuzhou Children's Hospital, Xuzhou Medical University, No 18 Sudi Road, Xuzhou 221000, China
| | - Yiyi Song
- Suzhou Medical College, Soochow University, 199 Ren-Ai Road, Suzhou 215123, China
| | - Xinxing Zhang
- Department of Pediatric Pulmonology, Children's Hospital of Soochow University, No 303, Jingde Road, Suzhou 215003, China
| | - Wenjing Gu
- Department of Pediatric Pulmonology, Children's Hospital of Soochow University, No 303, Jingde Road, Suzhou 215003, China
| | - Heting Dong
- Department of Pediatric Pulmonology, Children's Hospital of Soochow University, No 303, Jingde Road, Suzhou 215003, China
| | - Li Huang
- Department of Pediatric Pulmonology, Children's Hospital of Soochow University, No 303, Jingde Road, Suzhou 215003, China
| |
Collapse
|
2
|
Luo J, Wu T, Zhang J, Liang Z, Shao W, Wang D, Li L, Zuo D, Zhou J. D-mannose promotes diabetic wound healing through inhibiting advanced glycation end products formation in keratinocytes. Mol Med 2025; 31:15. [PMID: 39827347 PMCID: PMC11748336 DOI: 10.1186/s10020-025-01070-3] [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: 09/21/2024] [Accepted: 01/05/2025] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Diabetic chronic foot ulcers pose a significant therapeutic challenge around the world, resulting in adverse effects and complications in patients. D-mannose is enriched in cirtus peel and exerts beneficial effects among various diseases, especially against inflammation-related disorders. METHODS Here, we examined the potential effect of D-mannose during wound healing process in streptozotocin (STZ)-induced diabetes mice in vivo and by culturing keratinocytes under high glucose condition in vitro. The skin lesion healing was recorded in photos and evaluated by histochemical staining. What's more, the advanced glycation end products (AGEs) concentration in blood and mice skin was quantified. Apoptotic cells were assessed by flow cytometry and Western blotting. Inflammatory cytokines and cellular differential gene expression levels were measured by real-time PCR. The expression of the AMPK/Nrf2/HO-1 signaling-related molecules was determined by Western blotting. RESULTS We first found that topical supplementation of D-mannose remarkably improved skin wound healing in diabetes mice. Furthermore, both in vivo and in vitro experiments demonstrated that D-mannose reduced the AGEs generation. Mechanistically, D-mannose inhibited AGEs, then upregulated AMPK/Nrf2/HO-1 signaling in the context of high glucose to maintain keratinocyte normal functions, which positively influenced macrophage and fibroblast to accelerate diabetic wound healing. Noteworthily, these protective effects of D-mannose were abolished by the pretreatment with inhibitors of AGEs or AMPK. CONCLUSION As far as we know, this is the first study exploring the protective role of D-mannose on diabetic wound healing via topical supplementation. We find that D-mannose protects keratinocytes from high glucose stimulation via inhibition of AGEs formation as well as orchestrates inflammatory microenvironment in diabetic wounded skin, suggesting its supplementation as a potential therapy to promote refractory wound healing in diabetic patients.
Collapse
Affiliation(s)
- Jialiang Luo
- Institute of Molecular Immunology, Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Guangdong Province Key Laboratory of Immune Regulation and Immunotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, Guangdong, China
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, Guangdong, China
- Department of Dermatology, the Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Tianxing Wu
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jing Zhang
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Zhicheng Liang
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Weijie Shao
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Di Wang
- Department of Dermatology, Dermatology Hospital of Southern Medical University, Southern Medical University, Guangzhou, China
| | - Lei Li
- Institute of Molecular Immunology, Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Guangdong Province Key Laboratory of Immune Regulation and Immunotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Daming Zuo
- Institute of Molecular Immunology, Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Guangdong Province Key Laboratory of Immune Regulation and Immunotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| | - Jia Zhou
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
3
|
Mahdabi M, Mehrgan MS, Rajabi Islami H. Deciphering the impact of stickwater hydrolysate on growth performance, immune response, and IGF-1/PI3K/AKT/mTOR signaling pathway in Siberian sturgeon (Acipenser baerii) fingerlings. FISH PHYSIOLOGY AND BIOCHEMISTRY 2024; 50:2605-2618. [PMID: 39373813 DOI: 10.1007/s10695-024-01407-5] [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: 05/21/2024] [Accepted: 09/24/2024] [Indexed: 10/08/2024]
Abstract
A feeding trial lasting 56 days was carried out to assess how the inclusion of stickwater hydrolysate (SWH) in the diet of Siberian sturgeon (Acipenser baerii) fingerlings affected their growth performance, immunity, digestive enzyme activity, and gene expression linked to the IGF-1/PI3K/AKT/mTOR signaling pathway. Siberian sturgeon fingerlings were acclimatized and fed isonitrogenous, isoenergetic diets with varying SWH concentrations (0%, 0.5%, 1.5%, and 2.5%). Growth parameters, serum proteins, immunological and digestive enzyme activities, and gene expression levels were assessed post-trial. Results demonstrated that 0.5%, and 1.5% SWH treatments significantly improved weight gain, specific growth rate, feed conversion ratio, and protein efficiency ratio. Notably, these diets also elevated serum protein and plasma globulin levels, reduced albumin-to-globulin ratios, and enhanced lysozyme, myeloperoxidase (MPO) activities, and immunoglobulin (Ig) M levels, indicating an immunostimulatory effect. Digestive enzyme activities were markedly increased in the SWH groups, particularly at 1.5%. Gene expression analyses revealed upregulation of mtorc1, s6K, akt, pi3k, and igf1, with concurrent downregulation of 4e-bp1 in the muscle of fish, signifying activation of the IGF-1/PI3K/AKT/mTOR pathway, which is central to protein synthesis and muscle growth. In conclusion, SWH at appropriate levels significantly enhances growth, digestive efficiency, and immune function in Siberian sturgeon fingerlings, while also activating key metabolic pathways.
Collapse
Affiliation(s)
- Mahdad Mahdabi
- Department of Fisheries, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Mehdi Shamsaie Mehrgan
- Department of Fisheries, Science and Research Branch, Islamic Azad University, Tehran, Iran.
| | - Houman Rajabi Islami
- Department of Fisheries, Science and Research Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
4
|
Hong X, Huang S, Jiang H, Ma Q, Qiu J, Luo Q, Cao C, Xu Y, Chen F, Chen Y, Sun C, Fu H, Liu Y, Li C, Chen F, Qiu P. Alcohol-related liver disease (ALD): current perspectives on pathogenesis, therapeutic strategies, and animal models. Front Pharmacol 2024; 15:1432480. [PMID: 39669199 PMCID: PMC11635172 DOI: 10.3389/fphar.2024.1432480] [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: 05/14/2024] [Accepted: 10/22/2024] [Indexed: 12/14/2024] Open
Abstract
Alcohol-related liver disease (ALD) is a major cause of morbidity and mortality worldwide. It encompasses conditions such as fatty liver, alcoholic hepatitis, chronic hepatitis with liver fibrosis or cirrhosis, and hepatocellular carcinoma. Numerous recent studies have demonstrated the critical role of oxidative stress, abnormal lipid metabolism, endoplasmic reticulum stress, various forms of cell death (including apoptosis, necroptosis, and ferroptosis), intestinal microbiota dysbiosis, liver immune response, cell autophagy, and epigenetic abnormalities in the pathogenesis of ALD. Currently, abstinence, corticosteroids, and nutritional therapy are the traditional therapeutic interventions for ALD. Emerging therapies for ALD mainly include the blockade of inflammatory pathways, the promotion of liver regeneration, and the restoration of normal microbiota. Summarizing the advances in animal models of ALD will facilitate a more systematic investigation of the pathogenesis of ALD and the exploration of therapeutic targets. This review summarizes the latest insight into the pathogenesis and molecular mechanisms of ALD, as well as the pros and cons of ALD rodent models, providing a basis for further research on therapeutic strategies for ALD.
Collapse
Affiliation(s)
- Xiao Hong
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Shuo Huang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - He Jiang
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qing Ma
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jiang Qiu
- Department of Medicine, Hangzhou Normal University, Hangzhou, China
| | - Qihan Luo
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chunlu Cao
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yiyang Xu
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Fuzhe Chen
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yufan Chen
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chunfeng Sun
- The First People’s Hospital of Xiaoshan District, Xiaoshan Affiliated Hospital of Wenzhou Medical University, Hangzhou, China
| | - Haozhe Fu
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yiming Liu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Changyu Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Fangming Chen
- Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ping Qiu
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
5
|
Yang Y, Wang Z, Ge H, Wang B, Xing P, Wang N, Song Z, Lin Y, Hou X. Leptin signaling promotes milk fat synthesis via PI3K/AKT/mTOR/SREBP1 in mammary gland of dairy cow. J DAIRY RES 2024; 91:433-444. [PMID: 40040580 DOI: 10.1017/s002202992500010x] [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] [Indexed: 03/06/2025]
Abstract
Milk fat synthesis is tightly regulated by hormones and growth factors. Leptin is a versatile peptide hormone that exerts pleiotropic effects on metabolic pathways. In this study, we evaluated the expression and function of leptin and its long form receptor OB-Rb in dairy cow mammary tissues from different physiological stages and in cultured mammary epithelial cells. The results showed that the expression of leptin and OB-Rb were significantly higher in the mammary tissues of lactating cows as compared with dry cows, suggesting that they are related to milk component synthesis. In cultured dairy cow mammary epithelial cells, leptin treatment significantly increased OB-Rb expression and intracellular triacylglycerol content. Transcriptome analysis identified the difference in gene expression between leptin treated cells and control cells, and 317 differentially expressed genes were identified. Gene ontology and pathway mapping showed that lipid metabolism-related gene expression increased and signal transduction pathway-related genes were the most significantly enriched. Mechanistic studies showed that leptin stimulation enhanced sterol regulatory element-binding protein 1 expression via activating the phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) signaling pathway, which in turn up-regulated the expression of genes related to milk fat synthesis. Moreover, we found that fatty acid synthesis precursors, acetate and β-hydroxybutyrate, could positively regulate the expression of leptin and OB-Rb in bovine mammary epithelial cells, thereby potentially increasing milk fat synthesis. Our study provided novel evidence in the regulation of leptin on milk fat production in mammary glands of dairy cows, as well as experimental basis for artificial regulation of milk fat.
Collapse
Affiliation(s)
- Yang Yang
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China
| | - Zhihui Wang
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China
| | - Huiju Ge
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China
| | - Bo Wang
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China
| | - Pengfei Xing
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China
| | - Nan Wang
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China
| | - Zhiyi Song
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China
| | - Ye Lin
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China
- Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin 150030, China
| | - Xiaoming Hou
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China
| |
Collapse
|
6
|
Paudel D, Hao F, Goand UK, Tian S, Koehle AM, Nguyen LV, Tian Y, Patterson AD, Singh V. Elevated systemic total bile acids escalate susceptibility to alcohol-associated liver disease. iScience 2024; 27:110940. [PMID: 39398234 PMCID: PMC11467679 DOI: 10.1016/j.isci.2024.110940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 06/21/2024] [Accepted: 09/10/2024] [Indexed: 10/15/2024] Open
Abstract
Excessive alcohol consumption is a major global health problem. Individuals with alcoholic liver disease often exhibit elevated serum total bile acids (TBAs). Nevertheless, the extent to which high TBA contributes to alcohol-associated liver disease (AALD) remains elusive. To investigate this, wild-type mice were categorized into normal (nTBA) and high (hTBA) TBA groups. Both groups underwent chronic-binge ethanol feeding for 4 weeks, followed by additional weekly ethanol doses. Ethanol feeding worsened AALD in both male and female mice with elevated serum TBA, characterized by liver dysfunction and steatosis. Decreased hepatic expression of genes involved in mitochondrial β-oxidation and lipid transport in ethanol-fed hTBA mice suggests that altered fatty acid metabolism contributed to AALD. Our findings, which represent the first to link high serum TBA to increased AALD susceptibility, underscore the importance of proactive serum TBA screening as a valuable tool for identifying individuals at high risk of developing AALD.
Collapse
Affiliation(s)
- Devendra Paudel
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Fuhua Hao
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Umesh K. Goand
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Sangshan Tian
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Anthony M. Koehle
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Loi V. Nguyen
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Yuan Tian
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Andrew D. Patterson
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Vishal Singh
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, USA
- Center for Molecular Immunology and Infectious Disease, Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, USA
- One Health Microbiome Center, Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, USA
| |
Collapse
|
7
|
Ding Z, Wei Y, Dai J, Pan C, Yang L, Li Q, Zhang Y, Yan Q, Wu C, Li A, Lan Z, Liu S, Wang X. Deficiency of SDHC promotes metastasis by reprogramming fatty acid metabolism in colorectal cancer. J Transl Med 2024; 22:544. [PMID: 38844980 PMCID: PMC11157952 DOI: 10.1186/s12967-024-05361-x] [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: 02/20/2024] [Accepted: 05/31/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND Several studies have demonstrated a strong correlation between impaired Succinate dehydrogenase (SDH) function and the advancement of tumors. As a subunit of SDH, succinate dehydrogenase complex subunit C (SDHC) has been revealed to play tumor suppressive roles in several cancers, while its specific role in colorectal cancer (CRC) still needs further investigation. METHODS Online database were utilized to investigate the expression of SDHC in colorectal cancer and to assess its correlation with patient prognosis. Cell metastasis was assessed using transwell and wound healing assays, while tumor metastasis was studied in a nude mice model in vivo. Drug screening and RNA sequencing were carried out to reveal the tumor suppressor mechanism of SDHC. Triglycerides, neutral lipids and fatty acid oxidation were measured using the Triglyceride Assay Kit, BODIPY 493/503 and Colorimetric Fatty Acid Oxidation Rate Assay Kit, respectively. The expression levels of enzymes involved in fatty acid metabolism and the PI3K/AKT signaling pathway were determined by quantitative real-time PCR and western blot. RESULTS Downregulation of SDHC was found to be closely associated with a poor prognosis in CRC. SDHC knockdown promoted CRC metastasis both in vitro and in vivo. Through drug screening and Gene set enrichment analysis, it was discovered that SDHC downregulation was positively associated with the fatty acid metabolism pathways significantly. The effects of SDHC silencing on metastasis were reversed when fatty acid synthesis was blocked. Subsequent experiments revealed that SDHC silencing activated the PI3K/AKT signaling axis, leading to lipid accumulation by upregulating the expression of aldehyde dehydrogenase 3 family member A2 (ALDH3A2) and reduction of fatty acid oxidation rate by suppressing the expression of acyl-coenzyme A oxidase 1 (ACOX1) and carnitine palmitoyltransferase 1A (CPT1A). CONCLUSIONS SDHC deficiency could potentially enhance CRC metastasis by modulating the PI3K/AKT pathways and reprogramming lipid metabolism.
Collapse
Affiliation(s)
- Zhuoyu Ding
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yiyi Wei
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jingping Dai
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chaomin Pan
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Li Yang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qingyuan Li
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yue Zhang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qun Yan
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Changjie Wu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Aimin Li
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhixian Lan
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Side Liu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
- Pazhou Lab, Guangzhou, Guangdong, China.
| | - Xinke Wang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
8
|
Zhang Y, Zhang XY, Shi SR, Ma CN, Lin YP, Song WG, Guo SD. Natural products in atherosclerosis therapy by targeting PPARs: a review focusing on lipid metabolism and inflammation. Front Cardiovasc Med 2024; 11:1372055. [PMID: 38699583 PMCID: PMC11064802 DOI: 10.3389/fcvm.2024.1372055] [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: 01/17/2024] [Accepted: 04/09/2024] [Indexed: 05/05/2024] Open
Abstract
Inflammation and dyslipidemia are critical inducing factors of atherosclerosis. Peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors and control the expression of multiple genes that are involved in lipid metabolism and inflammatory responses. However, synthesized PPAR agonists exhibit contrary therapeutic effects and various side effects in atherosclerosis therapy. Natural products are structural diversity and have a good safety. Recent studies find that natural herbs and compounds exhibit attractive therapeutic effects on atherosclerosis by alleviating hyperlipidemia and inflammation through modulation of PPARs. Importantly, the preparation of natural products generally causes significantly lower environmental pollution compared to that of synthesized chemical compounds. Therefore, it is interesting to discover novel PPAR modulator and develop alternative strategies for atherosclerosis therapy based on natural herbs and compounds. This article reviews recent findings, mainly from the year of 2020 to present, about the roles of natural herbs and compounds in regulation of PPARs and their therapeutic effects on atherosclerosis. This article provides alternative strategies and theoretical basis for atherosclerosis therapy using natural herbs and compounds by targeting PPARs, and offers valuable information for researchers that are interested in developing novel PPAR modulators.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Endocrinology and Metabolism, Guiqian International General Hospital, Guiyang, China
| | - Xue-Ying Zhang
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, Weifang, China
| | - Shan-Rui Shi
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, Weifang, China
| | - Chao-Nan Ma
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, Weifang, China
| | - Yun-Peng Lin
- Department of General Surgery, Qixia Traditional Chinese Medicine Hospital in Shandong Province, Yantai, China
| | - Wen-Gang Song
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Shou-Dong Guo
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, Weifang, China
| |
Collapse
|
9
|
Shamsan E, Almezgagi M, Gamah M, Khan N, Qasem A, Chuanchuan L, Haining F. The role of PI3k/AKT signaling pathway in attenuating liver fibrosis: a comprehensive review. Front Med (Lausanne) 2024; 11:1389329. [PMID: 38590313 PMCID: PMC10999701 DOI: 10.3389/fmed.2024.1389329] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 03/11/2024] [Indexed: 04/10/2024] Open
Abstract
Excessive accumulation of extracellular matrix (ECM) components within the liver leads to a pathological condition known as liver fibrosis. Alcohol abuse, non-alcoholic fatty liver disease (NAFLD), autoimmune issues, and viral hepatitis cause chronic liver injury. Exploring potential therapeutic targets and understanding the molecular mechanisms involved in liver fibrosis are essential for the development of effective interventions. The goal of this comprehensive review is to explain how the PI3K/AKT signaling pathway contributes to the reduction of liver fibrosis. The potential of this pathway as a therapeutic target is investigated through a summary of results from in vivo and in vitro studies. Studies focusing on PI3K/AKT activation have shown a significant decrease in fibrosis markers and a significant improvement in liver function. The review emphasizes how this pathway may prevent ECM synthesis and hepatic stellate cell (HSC) activation, ultimately reducing the fibrotic response. The specific mechanisms and downstream effectors of the PI3K/AKT pathway in liver fibrosis constitute a rapidly developing field of study. In conclusion, the PI3K/AKT signaling pathway plays a significant role in attenuating liver fibrosis. Its complex role in regulating HSC activation and ECM production, demonstrated both in vitro and in vivo, underscores its potential as a effective therapeutic approach for managing liver fibrosis and slowing disease progression. A comprehensive review of this field provides valuable insights into its future developments and implications for clinical applications.
Collapse
Affiliation(s)
- Emad Shamsan
- College of Clinical Medicine, Qinghai University, Xining, China
- College of Medical Science, Taiz University, Taiz, Yemen
| | - Maged Almezgagi
- College of Clinical Medicine, Qinghai University, Xining, China
| | - Mohammed Gamah
- College of Clinical Medicine, Qinghai University, Xining, China
| | - Naveed Khan
- College of Clinical Medicine, Qinghai University, Xining, China
| | | | - Liu Chuanchuan
- College of Clinical Medicine, Qinghai University, Xining, China
- Qinghai University Affiliated Hospital, Xining, China
| | - Fan Haining
- College of Clinical Medicine, Qinghai University, Xining, China
- Qinghai University Affiliated Hospital, Xining, China
| |
Collapse
|
10
|
Li L, Kong L, Xu S, Wang C, Gu J, Luo H, Meng Q. FXR overexpression prevents hepatic steatosis through inhibiting AIM2 inflammasome activation in alcoholic liver disease. Hepatol Int 2024; 18:188-205. [PMID: 38183609 DOI: 10.1007/s12072-023-10621-x] [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: 08/30/2023] [Accepted: 11/22/2023] [Indexed: 01/08/2024]
Abstract
BACKGROUND AND PURPOSE Alcoholic liver disease (ALD), a metabolic liver disease caused by excessive alcohol consumption, has attracted increasing attention due to its high prevalence and mortality. Up to date, there is no effective and feasible treatment method for ALD. This study was to investigate whether Farnesoid X receptor (FXR, NR1H4) can alleviate ALD and whether this effect is mediated by inhibiting absent in melanoma 2 (AIM2) inflammasome activation. METHODS The difference in FXR expression between normal subjects and ALD patients was analyzed using the Gene Expression Omnibus (GEO) database. Lieber-DeCarli liquid diet with 5% ethanol (v/v) (EtOH) was adopted to establish the mouse ALD model. Liver histopathological changes and the accumulation of lipid droplets were assessed by H&E and Oil Red O staining. Quantitative real-time PCR, Western blotting analysis and immunofluorescence staining were utilized to evaluate the expression levels of related genes and proteins. DCFH-DA staining was adopted to visualize reactive oxidative species (ROS). RESULTS FXR was distinctly downregulated in liver tissues of patients with steatosis compared to normal livers using the GEO database, and in ethanol-induced AML-12 cellular steatosis model. FXR overexpression ameliorated hepatic lipid metabolism disorder and steatosis induced by ethanol by inhibiting the expression of genes involved in lipid synthesis and inducing the expression of genes responsible for lipid metabolism. Besides, FXR overexpression inhibited ethanol-induced AIM2 inflammasome activation and alleviated oxidative stress and ROS production during ethanol-induced hepatic steatosis. However, when FXR was knocked down, the results were completely opposite. CONCLUSIONS FXR attenuated lipid metabolism disorders and lipid degeneration in alcohol-caused liver injury and alleviated oxidative stress and inflammation by inhibiting AIM2 inflammasome activation.
Collapse
Affiliation(s)
- Lin Li
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, 9 West Section, Lvshun South Road, Dalian, 116044, China
| | - Lina Kong
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, 9 West Section, Lvshun South Road, Dalian, 116044, China
| | - Shuai Xu
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, 9 West Section, Lvshun South Road, Dalian, 116044, China
| | - Changyuan Wang
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, 9 West Section, Lvshun South Road, Dalian, 116044, China
| | - Jiangning Gu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Haifeng Luo
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Qiang Meng
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, 9 West Section, Lvshun South Road, Dalian, 116044, China.
| |
Collapse
|
11
|
Xiao Y, Yang Y, Xiong H, Dong G. The implications of FASN in immune cell biology and related diseases. Cell Death Dis 2024; 15:88. [PMID: 38272906 PMCID: PMC10810964 DOI: 10.1038/s41419-024-06463-6] [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: 06/13/2023] [Revised: 01/05/2024] [Accepted: 01/10/2024] [Indexed: 01/27/2024]
Abstract
Fatty acid metabolism, particularly fatty acid synthesis, is a very important cellular physiological process in which nutrients are used for energy storage and biofilm synthesis. As a key enzyme in the fatty acid metabolism, fatty acid synthase (FASN) is receiving increasing attention. Although previous studies on FASN have mainly focused on various malignancies, many studies have recently reported that FASN regulates the survival, differentiation, and function of various immune cells, and subsequently participates in the occurrence and development of immune-related diseases. However, few studies to date systematically summarized the function and molecular mechanisms of FASN in immune cell biology and related diseases. In this review, we discuss the regulatory effect of FASN on immune cells, and the progress in research on the implications of FASN in immune-related diseases. Understanding the function of FASN in immune cell biology and related diseases can offer insights into novel treatment strategies for clinical diseases.
Collapse
Affiliation(s)
- Yucai Xiao
- Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, 272067, Shandong, China
- Jining Key Laboratory of Immunology, Jining Medical University, Jining, 272067, Shandong, China
| | - Yonghong Yang
- Medical Research Center, Affiliated Hospital of Jining Medical University, Jining, 272007, Shandong, China
| | - Huabao Xiong
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, 272067, Shandong, China.
- Jining Key Laboratory of Immunology, Jining Medical University, Jining, 272067, Shandong, China.
| | - Guanjun Dong
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, 272067, Shandong, China.
- Jining Key Laboratory of Immunology, Jining Medical University, Jining, 272067, Shandong, China.
| |
Collapse
|
12
|
Hong JG, Carbajal Y, Trotman J, Glass M, Sclar V, Alter IL, Zhang P, Wang L, Chen L, Petitjean M, Friedman SL, DeRossi C, Chu J. Mannose Supplementation Curbs Liver Steatosis and Fibrosis in Murine MASH by Inhibiting Fructose Metabolism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.17.576067. [PMID: 38293175 PMCID: PMC10827199 DOI: 10.1101/2024.01.17.576067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) can progress to cirrhosis and liver cancer. There are no approved medical therapies to prevent or reverse disease progression. Fructose and its metabolism in the liver play integral roles in MASH pathogenesis and progression. Here we focus on mannose, a simple sugar, which dampens hepatic stellate cell activation and mitigates alcoholic liver disease in vitro and in vivo . In the well-validated FAT-MASH murine model, oral mannose supplementation improved both liver steatosis and fibrosis at low and high doses, whether administered either at the onset of the model ("Prevention") or at week 6 of the 12-week MASH regimen ("Reversal"). The in vivo anti-fibrotic effects of mannose supplementation were validated in a second model of carbon tetrachloride-induced liver fibrosis. In vitro human and mouse primary hepatocytes revealed that the anti-steatotic effects of mannose are dependent on the presence of fructose, which attenuates expression of ketohexokinase (KHK), the main enzyme in fructolysis. KHK is decreased with mannose supplementation in vivo and in vitro, and overexpression of KHK abrogated the anti-steatotic effects of mannose. Our study identifies mannose as a simple, novel therapeutic candidate for MASH that mitigates metabolic dysregulation and exerts anti-fibrotic effects.
Collapse
|
13
|
Liu Y, Xing L, Zhang Y, Liu X, Li T, Zhang S, Wei H, Li J. Mild Intermittent Cold Stimulation Affects Cardiac Substance Metabolism via the Neuroendocrine Pathway in Broilers. Animals (Basel) 2023; 13:3577. [PMID: 38003194 PMCID: PMC10668735 DOI: 10.3390/ani13223577] [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: 09/04/2023] [Revised: 11/08/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023] Open
Abstract
This study aimed to investigate the impact of cold adaptation on the neuroendocrine and cardiac substance metabolism pathways in broilers. The broilers were divided into the control group (CC), cold adaptation group (C3), and cold-stressed group (C9), and experimental period was divided into the training period (d 1-35), recovery period (d 36-43), and cold stress period (d 43-44). During the training period, the CC group was reared at ambient temperature, while C3 and C9 groups were reared at 3 °C and 9 °C lower than the ambient temperature, respectively, for 5 h/d at 1 d intervals. During the recovery period, all the groups were maintained at 20 °C. Lastly, during the cold stress period, the groups were divided into two sub-groups, and each sub-group was placed at 10 °C for 12 h (Y12) or 24 h (Y24) for acute cold stimulation. The blood, hypothalamic, and cardiac tissues samples were obtained from all the groups during the training, recovery, and acute stress periods. The results revealed that the transcription of calcium voltage-gated channel subunit alpha 1 C (CACNAIC) was increased in the hypothalamic tissues of the C3 group (p < 0.05). Moreover, compared to the CC group, the serum norepinephrine (NE) was increased in the C9 group (p < 0.05), but insulin (INS) was decreased in the C9 group (p < 0.05). In addition, the transcription of the phosphoinositide-3 kinase (PI3K), protein kinase B (Akt), mammalian target of rapamycin (mTOR), SREBP1c, FASN, ACC1, and SCD genes was down-regulated in the C3 and C9 groups (p < 0.05); however, their expression increased in the C3 and C9 groups after acute cold stimulation (p < 0.05). Compared to the CC group, the transcription of forkhead box O1 (FoxO1), PEPCK, G6Pase, GLUT1, HK1, PFK, and LDHB genes was up-regulated in the C3 and C9 groups (p < 0.05. Furthermore, compared to the CC and C9 groups, the protein and mRNA expressions of heat shock protein (HSP) 70 and HSP90 were significantly increased in the C3 group (p < 0.05). These results indicate that intermittent cold training can enhance cold stress tolerance in broilers by regulating their neuroendocrine and cardiac substance metabolism pathways.
Collapse
Affiliation(s)
- Yuanyuan Liu
- College of Life Science, Northeast Agricultural University, Harbin 150030, China; (Y.L.); (L.X.); (Y.Z.); (X.L.); (T.L.); (S.Z.)
| | - Lu Xing
- College of Life Science, Northeast Agricultural University, Harbin 150030, China; (Y.L.); (L.X.); (Y.Z.); (X.L.); (T.L.); (S.Z.)
| | - Yong Zhang
- College of Life Science, Northeast Agricultural University, Harbin 150030, China; (Y.L.); (L.X.); (Y.Z.); (X.L.); (T.L.); (S.Z.)
| | - Xiaotao Liu
- College of Life Science, Northeast Agricultural University, Harbin 150030, China; (Y.L.); (L.X.); (Y.Z.); (X.L.); (T.L.); (S.Z.)
| | - Tingting Li
- College of Life Science, Northeast Agricultural University, Harbin 150030, China; (Y.L.); (L.X.); (Y.Z.); (X.L.); (T.L.); (S.Z.)
| | - Shijie Zhang
- College of Life Science, Northeast Agricultural University, Harbin 150030, China; (Y.L.); (L.X.); (Y.Z.); (X.L.); (T.L.); (S.Z.)
| | - Haidong Wei
- College of Life Science, Northeast Agricultural University, Harbin 150030, China; (Y.L.); (L.X.); (Y.Z.); (X.L.); (T.L.); (S.Z.)
| | - Jianhong Li
- College of Life Science, Northeast Agricultural University, Harbin 150030, China; (Y.L.); (L.X.); (Y.Z.); (X.L.); (T.L.); (S.Z.)
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin 150030, China
| |
Collapse
|
14
|
Yang Y, Liu C, Zhang C, Xu Z, Zhang L, Cui Y, Wang C, Lin Y, Hou X. Acetate Upregulates GPR43 Expression and Function via PI3K-AKT-SP1 Signaling in Mammary Epithelial Cells during Milk Fat Synthesis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:16003-16015. [PMID: 37870996 DOI: 10.1021/acs.jafc.3c00965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
This study investigated the mechanism underlying acetate-induced orphan G-protein-coupled receptor 43 (GPR43) expression and milk fat production. The mammary epithelial cells of dairy cows were treated with acetate, and the effects of GPR43 on acetate uptake and the expression of lipogenesis-related genes were determined by gas chromatography and quantitative polymerase chain reaction (qPCR), respectively. RNAi, inhibitor treatment, and luciferase assay were used to determine the effect of phosphoinositide 3-kinase-protein kinase B-specificity protein 1 (PI3K-AKT-SP1) signaling on acetate-induced GPR43 expression and function. The results showed that GPR43 was highly expressed in lactating cow mammary tissues, which was related to milk fat synthesis. 12 mM acetate significantly increased the GPR43 expression in mammary epithelial cells of dairy cows. In acetate-treated cells, GPR43 overexpression significantly increased the cellular uptake of acetate, the intracellular triacylglycerol (TAG) content, and acetate-induced lipogenesis gene expression. Acetate activated PI3K-AKT signaling and promoted SP1 translocation from the cytosol into the nucleus, where SP1 bound to the GPR43 promoter and upregulated GPR43 transcription. Moreover, the activation of PI3K-AKT-SP1 by acetate facilitated the trafficking of GPR43 from the cytosol to the plasma membrane. In conclusion, acetate upregulated GPR43 expression and function via PI3K-AKT-SP1 signaling in mammary epithelial cells, thereby increasing milk fat synthesis. These results provide an experimental strategy for improving milk lipid synthesis, which is important to the dairy industry.
Collapse
Affiliation(s)
- Yang Yang
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China
| | - Chuanping Liu
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China
| | - Caiyan Zhang
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China
| | - Ziru Xu
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China
| | - Li Zhang
- Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin 150030, China
| | - Yingjun Cui
- Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin 150030, China
| | - Chunmei Wang
- Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin 150030, China
| | - Ye Lin
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China
- Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin 150030, China
| | - Xiaoming Hou
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China
| |
Collapse
|
15
|
Qiu F, Zeng R, Li D, Ye T, Xu W, Wang X, Yan X, Li H, Hu X. Establishment and bioinformatics evaluation of the ethanol combined with palmitic acid-induced mouse hepatocyte AFLD model (the Hu-Qiu Model). Heliyon 2023; 9:e19359. [PMID: 37681138 PMCID: PMC10481297 DOI: 10.1016/j.heliyon.2023.e19359] [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: 12/21/2022] [Revised: 08/15/2023] [Accepted: 08/20/2023] [Indexed: 09/09/2023] Open
Abstract
Chronic alcoholic liver disease has brought great harm to human health. Alcoholic fatty liver disease is the first stage in the progression of all chronic alcoholic liver diseases. At present, there is no cell model that fully matches the etiology (high-fat diet + alcohol) of human alcoholic fatty liver disease. We used 100 mM ethanol +6.25 μM PA to establish the ethanol combined with PA-induced mouse hepatocyte AFLD model (EP-AFLD hepatocyte model) and performed the RNA-seq transcriptome sequencing. Through bioinformatics analysis and comparison, we discovered that the EP-AFLD hepatocyte model was more suitable for studying the pathological mechanism of AFLD than the mouse AFLD hepatocyte model induced by ethanol alone. And through bioinformatics analysis, we further discovered that 77 genes from the differential expression gene set of EP-AFLD hepatocyte model were engaged in the pathological process of mouse AFLD and 40 genes were involved in the pathogenesis of both mouse AFLD and human AFLD. In this study, a novel mouse hepatocyte AFLD model was successfully established by combining ethanol and PA, which can be used to study the molecular mechanism of the pathogenesis of AFLD in mice or humans. This study will provide a brand-new in vitro experimental platform for the in-depth study of AFLD pathogenesis and the screening of AFLD therapeutic drugs.
Collapse
Affiliation(s)
| | | | - Du Li
- Department of Biology, School of Basic Medical Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Tingjie Ye
- Department of Biology, School of Basic Medical Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Wei Xu
- Department of Biology, School of Basic Medical Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xiaoling Wang
- Department of Biology, School of Basic Medical Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xiaofeng Yan
- Department of Biology, School of Basic Medical Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Hua Li
- Department of Biology, School of Basic Medical Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xudong Hu
- Department of Biology, School of Basic Medical Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| |
Collapse
|
16
|
Zhang CY, Liu S, Yang M. Antioxidant and anti-inflammatory agents in chronic liver diseases: Molecular mechanisms and therapy. World J Hepatol 2023; 15:180-200. [PMID: 36926234 PMCID: PMC10011909 DOI: 10.4254/wjh.v15.i2.180] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 11/30/2022] [Accepted: 02/07/2023] [Indexed: 02/24/2023] Open
Abstract
Chronic liver disease (CLD) is a continuous process that causes a reduction of liver function lasting more than six months. CLD includes alcoholic liver disease (ALD), non-alcoholic fatty liver disease (NAFLD), chronic viral infection, and autoimmune hepatitis, which can lead to liver fibrosis, cirrhosis, and cancer. Liver inflammation and oxidative stress are commonly associated with the development and progression of CLD. Molecular signaling pathways such as AMP-activated protein kinase (AMPK), C-Jun N-terminal kinase, and peroxisome proliferator-activated receptors (PPARs) are implicated in the pathogenesis of CLD. Therefore, antioxidant and anti-inflammatory agents from natural products are new potent therapies for ALD, NAFLD, and hepatocellular carcinoma (HCC). In this review, we summarize some powerful products that can be potential applied in all the stages of CLD, from ALD/NAFLD to HCC. The selected agents such as β-sitosterol, curcumin, genistein, and silymarin can regulate the activation of several important molecules, including AMPK, Farnesoid X receptor, nuclear factor erythroid 2-related factor-2, PPARs, phosphatidylinositol-3-kinase, and lysyl oxidase-like proteins. In addition, clinical trials are undergoing to evaluate their efficacy and safety.
Collapse
Affiliation(s)
- Chun-Ye Zhang
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, United States
| | - Shuai Liu
- The First Affiliated Hospital, Zhejiang University, Hangzhou 310006, Zhejiang Province, China
| | - Ming Yang
- Department of Surgery, University of Missouri, Columbia, MO 65211, United States.
| |
Collapse
|
17
|
Nguyen Huu T, Park J, Zhang Y, Duong Thanh H, Park I, Choi JM, Yoon HJ, Park SC, Woo HA, Lee SR. The Role of Oxidative Inactivation of Phosphatase PTEN and TCPTP in Fatty Liver Disease. Antioxidants (Basel) 2023; 12:antiox12010120. [PMID: 36670982 PMCID: PMC9854873 DOI: 10.3390/antiox12010120] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/29/2022] [Accepted: 12/30/2022] [Indexed: 01/05/2023] Open
Abstract
Alcoholic liver disease (ALD) and nonalcoholic fatty liver disease (NAFLD) are becoming increasingly prevalent worldwide. Despite the different etiologies, their spectra and histological feature are similar, from simple steatosis to more advanced stages such as steatohepatitis, fibrosis, cirrhosis, and hepatocellular carcinoma. Studies including peroxiredoxin knockout models revealed that oxidative stress is crucial in these diseases, which present as consequences of redox imbalance. Protein tyrosine phosphatases (PTPs) are a superfamily of enzymes that are major targets of reactive oxygen species (ROS) because of an oxidation-susceptible nucleophilic cysteine in their active site. Herein, we review the oxidative inactivation of two tumor suppressor PTPs, phosphatase and tensin homolog deleted on chromosome 10 (PTEN) and T-cell protein tyrosine phosphatase (TCPTP), and their contribution to the pathogenicity of ALD and NAFLD, respectively. This review might provide a better understanding of the pathogenic mechanisms of these diseases and help develop new therapeutic strategies to treat fatty liver disease.
Collapse
Affiliation(s)
- Thang Nguyen Huu
- Department of Biochemistry, Department of Biomedical Sciences, Research Center for Aging and Geriatrics, Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
- BioMedical Sciences Graduate Program (BMSGP), Chonnam National University Medical School, Hwasun 58 128, Republic of Korea
| | - Jiyoung Park
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 120-750, Republic of Korea
| | - Ying Zhang
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Hien Duong Thanh
- BioMedical Sciences Graduate Program (BMSGP), Chonnam National University Medical School, Hwasun 58 128, Republic of Korea
- Department of Anatomy, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| | - Iha Park
- Department of Biochemistry, Department of Biomedical Sciences, Research Center for Aging and Geriatrics, Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| | - Jin Myung Choi
- Department of Biochemistry, Department of Biomedical Sciences, Research Center for Aging and Geriatrics, Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| | - Hyun Joong Yoon
- Department of Biochemistry, Department of Biomedical Sciences, Research Center for Aging and Geriatrics, Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| | - Sang Chul Park
- The Future Life and Society Research Center, Advanced Institute of Aging Science, Chonnam National University, Gwangju 61469, Republic of Korea
| | - Hyun Ae Woo
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 120-750, Republic of Korea
| | - Seung-Rock Lee
- Department of Biochemistry, Department of Biomedical Sciences, Research Center for Aging and Geriatrics, Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
- Correspondence: ; Tel.: +82-61-379-2775; Fax: +82-61-379-2782
| |
Collapse
|
18
|
Daucosterol Alleviates Alcohol-Induced Hepatic Injury and Inflammation through P38/NF-κB/NLRP3 Inflammasome Pathway. Nutrients 2023; 15:nu15010223. [PMID: 36615880 PMCID: PMC9823995 DOI: 10.3390/nu15010223] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/23/2022] [Accepted: 12/28/2022] [Indexed: 01/04/2023] Open
Abstract
Alcoholic liver disease (ALD) is caused by chronic excessive alcohol consumption, which leads to inflammation, oxidative stress, lipid accumulation, liver fibrosis/cirrhosis, and even liver cancer. However, there are currently no effective drugs for ALD. Herein, we report that a natural phytosterol Daucosterol (DAU) can effectively protect against liver injury caused by alcohol, which plays anti-inflammatory and antioxidative roles in many chronic inflammatory diseases. Our results demonstrate that DAU ameliorates liver inflammation induced by alcohol through p38/nuclear factor kappa B (NF-κB)/NOD-like receptor protein-3 (NLRP3) inflammasome pathway. Briefly, DAU decreases NF-κB nuclear translocation and inhibits NLRP3 activation by decreasing p38 phosphorylation. At the same time, DAU also protects against hepatic oxidative stress and lipid accumulation. In conclusion, our research provides a new clue about the protective effects of naturally active substances on ALD.
Collapse
|