1
|
Zhang L, Liu S, Zhao Q, Liu X, Zhang Q, Liu M, Zhao W. The role of ubiquitination and deubiquitination in the pathogenesis of non-alcoholic fatty liver disease. Front Immunol 2025; 16:1535362. [PMID: 40292292 PMCID: PMC12021615 DOI: 10.3389/fimmu.2025.1535362] [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] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 03/19/2025] [Indexed: 04/30/2025] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is one of the most common chronic liver diseases and is closely associated with metabolic abnormalities. The causes of NAFLD are exceedingly complicated, and it is known that a variety of signaling pathways, endoplasmic reticulum stress, and mitochondrial dysfunction play a role in the pathogenesis of NAFLD. Recent studies have shown that ubiquitination and deubiquitination are involved in the regulation of the NAFLD pathophysiology. Protein ubiquitination is a dynamic and diverse post-translational alteration that affects various cellular biological processes. Numerous disorders, including NAFLD, exhibit imbalances in ubiquitination and deubiquitination. To highlight the significance of this post-translational modification in the pathogenesis of NAFLD and to aid in the development of new therapeutic approaches for the disease, we will discuss the role of enzymes involved in the processes of ubiquitination and deubiquitination, specifically E3 ubiquitin ligases and deubiquitinating enzymes that are important in the regulation of NAFLD.
Collapse
Affiliation(s)
- Lihui Zhang
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
- Collaborative Innovation Center of Prevention and Treatment of Major Diseases by Chinese and Western Medicine, Zhengzhou, Henan, China
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Zhengzhou, Henan, China
| | - Sutong Liu
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
- Collaborative Innovation Center of Prevention and Treatment of Major Diseases by Chinese and Western Medicine, Zhengzhou, Henan, China
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Zhengzhou, Henan, China
| | - Qing Zhao
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Xiaoyan Liu
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Qiang Zhang
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Minghao Liu
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
- Collaborative Innovation Center of Prevention and Treatment of Major Diseases by Chinese and Western Medicine, Zhengzhou, Henan, China
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Zhengzhou, Henan, China
| | - Wenxiao Zhao
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
- Collaborative Innovation Center of Prevention and Treatment of Major Diseases by Chinese and Western Medicine, Zhengzhou, Henan, China
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Zhengzhou, Henan, China
| |
Collapse
|
2
|
Zhang Y, Yang J, Min J, Huang S, Li Y, Liu S. The emerging role of E3 ubiquitin ligases and deubiquitinases in metabolic dysfunction-associated steatotic liver disease. J Transl Med 2025; 23:368. [PMID: 40133964 PMCID: PMC11938720 DOI: 10.1186/s12967-025-06255-2] [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: 10/23/2024] [Accepted: 02/17/2025] [Indexed: 03/27/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most common chronic liver disease worldwide, with a prevalence as high as 32.4%. MASLD encompasses a spectrum of liver pathologies, ranging from steatosis to metabolic dysfunction-associated steatohepatitis (MASH), fibrosis, and, in some cases, progression to end-stage liver disease (cirrhosis and hepatocellular carcinoma). A comprehensive understanding of the pathogenesis of this highly prevalent liver disease may facilitate the identification of novel targets for the development of improved therapies. E3 ubiquitin ligases and deubiquitinases (DUBs) are key regulatory components of the ubiquitin‒proteasome system (UPS), which plays a pivotal role in maintaining intracellular protein homeostasis. Emerging evidence implicates that aberrant expression of E3 ligases and DUBs is involved in the progression of MASLD. Here, we review abnormalities in E3 ligases and DUBs by (1) discussing their targets, mechanisms, and functions in MASLD; (2) summarizing pharmacological interventions targeting these enzymes in preclinical and clinical studies; and (3) addressing challenges and future therapeutic strategies. This review synthesizes current evidence to highlight the development of novel therapeutic strategies based on the UPS for MASLD and progressive liver disease.
Collapse
Affiliation(s)
- Yu Zhang
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, CSU-Sinocare Research Center for Nutrition and Metabolic Health, Furong Laboratory, Changsha, Hunan, 410011, China
| | - Jiahui Yang
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, CSU-Sinocare Research Center for Nutrition and Metabolic Health, Furong Laboratory, Changsha, Hunan, 410011, China
| | - Jiali Min
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, CSU-Sinocare Research Center for Nutrition and Metabolic Health, Furong Laboratory, Changsha, Hunan, 410011, China
| | - Shan Huang
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, CSU-Sinocare Research Center for Nutrition and Metabolic Health, Furong Laboratory, Changsha, Hunan, 410011, China
| | - Yuchen Li
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, CSU-Sinocare Research Center for Nutrition and Metabolic Health, Furong Laboratory, Changsha, Hunan, 410011, China
| | - Shanshan Liu
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, CSU-Sinocare Research Center for Nutrition and Metabolic Health, Furong Laboratory, Changsha, Hunan, 410011, China.
| |
Collapse
|
3
|
Ye L, Yao Z, Xuan Q, Liu Q, Bo T. The impact of sleeve gastrectomy on MASH development by regulating the composition of gut microbiota and metabolic homeostasis. Biochem Biophys Res Commun 2025; 752:151466. [PMID: 39938449 DOI: 10.1016/j.bbrc.2025.151466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 01/21/2025] [Accepted: 02/07/2025] [Indexed: 02/14/2025]
Abstract
The prevalence of metabolic dysfunction-associated steatohepatitis (MASH) is increasing annually, which is a global public health issue. Although clinical trials are lacking, observational studies indicate that bariatric surgery can alleviate the progression of MASH. Here, we performed sleeve gastrectomy (SG) and Sham surgery on 8-week-old mice, and then fed a AMLN diet for 24 weeks to construct a diet-inducted MASH mice model after 4-week post-surgery recovery. Applying a multi-omics approach combining metagenomics, metabolomics, and transcriptomics, we found that SG prevents the development of hepatic steatosis, inflammation, and fibrosis in MASH mice not only by significantly altering the structure of gut microbiota including s_Akkermansia muciniphila, s_Alistiples dispar, g_Helicobacter and s_uc_Oscillospiraceae, but also by modulating the levels of serum metabolites including l-arginine and taurocholic acid (TCA). These results suggest that SG and the alteration of gut microbiota and its related serum metabolites can be served as the effective therapeutic strategies for MASH.
Collapse
Affiliation(s)
- Lingxi Ye
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Jinan, Shandong, 250021, China; Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Zhenyu Yao
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Jinan, Shandong, 250021, China; Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Qiuhui Xuan
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Jinan, Shandong, 250021, China; Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Qiaoran Liu
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, 250021, China; Key Laboratory of Metabolism and Gastrointestinal Tumor, the First Affiliated Hospital of Shandong First Medical University, China; Key Laboratory of Laparoscopic Technology, the First Affiliated Hospital of Shandong First Medical University, China.
| | - Tao Bo
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Jinan, Shandong, 250021, China; Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China.
| |
Collapse
|
4
|
Liu Y, Qian M, Li Y, Dong X, Wu Y, Yuan T, Ma J, Yang B, Zhu H, He Q. The ubiquitin-proteasome system: A potential target for the MASLD. Acta Pharm Sin B 2025; 15:1268-1280. [PMID: 40370547 PMCID: PMC12069246 DOI: 10.1016/j.apsb.2025.01.010] [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: 06/14/2024] [Revised: 09/20/2024] [Accepted: 12/20/2024] [Indexed: 05/16/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), the most prevalent chronic liver condition globally, lacks adequate and effective therapeutic remedies in clinical practice. Recent studies have increasingly highlighted the close connection between the ubiquitin-proteasome system (UPS) and the progression of MASLD. This relationship is crucial for understanding the disease's underlying mechanism. As a sophisticated process, the UPS govern protein stability and function, maintaining protein homeostasis, thus influencing a multitude of elements and biological events of eukaryotic cells. It comprises four enzyme families, namely, ubiquitin-activating enzymes (E1), ubiquitin-conjugating enzymes (E2), ubiquitin-protein ligases (E3), and deubiquitinating enzymes (DUBs). This review aims to delve into the array of pathways and therapeutic targets implicated in the ubiquitination within the pathogenesis of MASLD. Therefore, this review unveils the role of ubiquitination in MASLD while spotlighting potential therapeutic targets within the context of this disease.
Collapse
Affiliation(s)
- Yue Liu
- Institute of Pharmacology & Toxicology, Zhejiang Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Meijia Qian
- Institute of Pharmacology & Toxicology, Zhejiang Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Hangzhou Zhongmei Huadong Pharmaceut Co., Ltd., Hangzhou 310011, China
| | - Yonghao Li
- Institute of Pharmacology & Toxicology, Zhejiang Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xin Dong
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Yulian Wu
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Tao Yuan
- Institute of Pharmacology & Toxicology, Zhejiang Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jian Ma
- Center for Drug Safety Evaluation and Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Bo Yang
- Institute of Pharmacology & Toxicology, Zhejiang Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- School of Medicine, Hangzhou City University, Hangzhou 310015, China
| | - Hong Zhu
- Institute of Pharmacology & Toxicology, Zhejiang Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Engineering Research Center of Innovative Anticancer Drugs, Ministry of Education, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou 310058, China
| | - Qiaojun He
- Institute of Pharmacology & Toxicology, Zhejiang Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| |
Collapse
|
5
|
Peters N, Kanngießer S, Pajonk O, Salazar Claros R, Hubbe P, Mogk A, Schuck S. Reprograming of the ubiquitin ligase Ubr1 by intrinsically disordered Roq1 through cooperating multifunctional motifs. EMBO J 2025; 44:1774-1803. [PMID: 39920309 PMCID: PMC11914429 DOI: 10.1038/s44318-025-00375-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 01/22/2025] [Accepted: 01/24/2025] [Indexed: 02/09/2025] Open
Abstract
One way cells control the speed and specificity of protein degradation is by regulating the activity of ubiquitin ligases. Upon proteotoxic stress in yeast, the intrinsically disordered protein Roq1 binds the ubiquitin ligase Ubr1 as a pseudosubstrate, thereby modulating the degradation of substrates of the N-degron pathway and promoting the elimination of misfolded proteins. The mechanism underlying this reprograming of Ubr1 is unknown. Here, we show that Roq1 controls Ubr1 by means of two cooperating multifunctional motifs. The N-terminal arginine and a short hydrophobic motif of Roq1 interact with Ubr1 as part of a heterobivalent binding mechanism. Via its N-terminal arginine, Roq1 regulates the ubiquitination of various N-degron substrates and folded proteins. Via its hydrophobic motif, Roq1 accelerates the ubiquitination of misfolded proteins. These findings reveal how a small, intrinsically disordered protein with a simple architecture engages parallel channels of communication to reprogram a functionally complex ubiquitin ligase.
Collapse
Affiliation(s)
- Niklas Peters
- Heidelberg University Biochemistry Center, 69120, Heidelberg, Germany
| | | | - Oliver Pajonk
- Heidelberg University Biochemistry Center, 69120, Heidelberg, Germany
| | - Rafael Salazar Claros
- Heidelberg University Biochemistry Center, 69120, Heidelberg, Germany
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK
| | - Petra Hubbe
- Heidelberg University Biochemistry Center, 69120, Heidelberg, Germany
| | - Axel Mogk
- Center for Molecular Biology of Heidelberg University, 69120, Heidelberg, Germany
| | - Sebastian Schuck
- Heidelberg University Biochemistry Center, 69120, Heidelberg, Germany.
| |
Collapse
|
6
|
Shi L, Li Z, Ma X, Wang J, Wu Y, Zhu Y, Wang Y, Yang Y, Luo M, Li J, Sun X, He S. Effects of ultra-processed foods on the liver: insights from gut microbiome and metabolomics studies in rats. Front Nutr 2025; 11:1503879. [PMID: 39912061 PMCID: PMC11794082 DOI: 10.3389/fnut.2024.1503879] [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: 09/29/2024] [Accepted: 12/23/2024] [Indexed: 02/07/2025] Open
Abstract
Purpose High consumption of Ultra-processed foods (UPF) have been identified as a potential risk factor for Non-alcoholic fatty liver disease (NAFLD). Nevertheless, there is limited empirical evidence regarding the impact of UPF, which are typical combination of processed foods, on liver health through alterations in gut microbiota and metabolic processes. We aim to examine the potential impact of UPF on liver health and to explore the role of gut microbiota and metabolites. Methods This study used Sprague-Dawley rats to mimic modern UPF diets for 90 days. Some serum biochemical indices, inflammatory factors, oxidative stress markers, hematoxylin-eosin (HE) staining of the liver, 16S ribosomal RNA (rRNA) and Liquid chromatography-mass spectrometry (LC-MS) of rat feces were detected. Results The UPF diet-induced simple steatosis of the liver in rats without affecting the levels of IL-6, GSH, MDA, and SOD. Additionally, it modified the gut microbiota, increasing potentially harmful bacteria, such as norank_f__Desulfovibrionaceae and Staphylococcus, while also elevating the relative abundance of potentially beneficial bacteria, including Dubosiella and Allobaculum. Furthermore, the consumption of UPF led to a metabolomic disorder characterized by disruptions in the sphingolipid signaling pathway, sulfur relay system, and arachidonic acid metabolism. Conclusion In conclusion, the findings of this study indicate that the consumption of UPF influences the development of simple hepatic steatosis, potentially through alterations in gut microbiota and metabolomics.
Collapse
Affiliation(s)
- Liping Shi
- Department of Epidemiology and Health Statistics, School of Public Health, Ningxia Medical University, Yinchuan, China
| | - Zhuoyuan Li
- Department of Epidemiology and Health Statistics, School of Public Health, Ningxia Medical University, Yinchuan, China
| | - Xiaojun Ma
- Department of Epidemiology and Health Statistics, School of Public Health, Ningxia Medical University, Yinchuan, China
| | - Junru Wang
- Department of Epidemiology and Health Statistics, School of Public Health, Ningxia Medical University, Yinchuan, China
| | - Yueping Wu
- Department of Epidemiology and Health Statistics, School of Public Health, Ningxia Medical University, Yinchuan, China
| | - Yongbin Zhu
- Department of Epidemiology and Health Statistics, School of Public Health, Ningxia Medical University, Yinchuan, China
| | - Yanrong Wang
- Department of Epidemiology and Health Statistics, School of Public Health, Ningxia Medical University, Yinchuan, China
| | - Yue Yang
- Department of Epidemiology and Health Statistics, School of Public Health, Ningxia Medical University, Yinchuan, China
| | - Minxiu Luo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Jiangping Li
- Department of Epidemiology and Health Statistics, School of Public Health, Ningxia Medical University, Yinchuan, China
| | - Xian Sun
- Department of Epidemiology and Health Statistics, School of Public Health, Ningxia Medical University, Yinchuan, China
- Key Laboratory of Environmental Factors and Chronic Disease Control, Ningxia Medical University, Yinchuan, China
| | - Shulan He
- Department of Epidemiology and Health Statistics, School of Public Health, Ningxia Medical University, Yinchuan, China
- Key Laboratory of Environmental Factors and Chronic Disease Control, Ningxia Medical University, Yinchuan, China
| |
Collapse
|
7
|
Shen H, Zhou L, Yang Y, Shu H, Wu D, Yang S, Xie L, Yang L, Tian S, Zhang X, Ma R, Jiang L, Jiang M, Zhang H, Wang Y, Zhang H, Gao S, Xu L, Wang H. The gut microbiota-produced vitamin B6 mitigates alcohol-associated liver disease by attenuating hepatic oxidative stress damage. Hepatol Commun 2025; 9:e0599. [PMID: 39670862 PMCID: PMC11637752 DOI: 10.1097/hc9.0000000000000599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 09/22/2024] [Indexed: 12/14/2024] Open
Abstract
BACKGROUND Alcohol-associated liver disease (ALD) is a major clinical issue characterized by progressive stages, including hepatic steatosis, liver fibrosis, cirrhosis, and HCC. Patients with long-term chronic alcoholism often present with gut microbiota dysbiosis and reduced plasma levels of vitamin B6. This study aimed to verify that gut microbiota disruption in ALD significantly contributes to reduced in vivo production of vitamin B6 and to investigate the role of this reduction in the pathogenesis of ALD. METHODS The ALD was investigated utilizing the Gao-binge mouse model. Fecal microbial composition was analyzed in pair-fed mice and ALD mice to identify alcohol-induced functional changes in the microbiota. Additionally, liver protein expression profiles and liver and plasma metabolomic profiles were characterized to elucidate the role of vitamin B6 in ALD pathogenesis through integrated proteomic and metabolomic analyses. The findings were further validated using animal models and clinical patient samples. RESULTS Alcohol consumption disrupted the gut microbiota in the mice, impairing the vitamin B6 synthesis by intestinal microorganisms. Vitamin B6 deficiency aggravated the disorder of amino acid metabolism in the liver and inhibited ornithine aminotransferase expression, thereby worsening oxidative stress damage. In patients with ALD, significant disturbances of gut microbiota were observed, along with decreased intestinal vitamin B6 levels, which were negatively correlated with serum biochemical markers. CONCLUSIONS The imbalance of gut microbiota in ALD mice reduces vitamin B6 synthesis, which affects amino acid metabolism and glutathione synthesis in the liver, thereby exacerbating ALD. These findings suggest that vitamin B6 may play a critical protective role in ALD progression by regulating amino acid metabolism.
Collapse
Affiliation(s)
- Haiyuan Shen
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, Hefei, China
| | - Liangliang Zhou
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
- Department of Infectious Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yuanru Yang
- Department of Blood Transfusion, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Hang Shu
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
| | - Dongqing Wu
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
| | - Simin Yang
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
| | - Linxi Xie
- School of Basic Medical Science, Anhui Medical University, Hefei, China
| | - Lei Yang
- School of Basic Medical Science, Anhui Medical University, Hefei, China
| | - Shanfei Tian
- School of Basic Medical Science, Anhui Medical University, Hefei, China
| | - Xinru Zhang
- School of Basic Medical Science, Anhui Medical University, Hefei, China
| | - Rui Ma
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, Hefei, China
| | - Ling Jiang
- Department of Nephropathy, the First Affiliated Hospital, Anhui Medical University, Hefei, China
| | - Man Jiang
- Department of Neurology, Mengcheng First People’s Hospital, Bozhou, China
| | - Hao Zhang
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
| | - Yan Wang
- School of Health Service Management, Anhui Medical University, Hefei, China
| | - Hejiao Zhang
- Department of Gastroenterology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Shan Gao
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei, China
| | - Long Xu
- School of Basic Medical Science, Anhui Medical University, Hefei, China
| | - Hua Wang
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
| |
Collapse
|
8
|
Jiang Z, Zhang C, Yu X, Wang K, Sang Z, Gong W, Zhang Q, Meng X, Qin L, Zhao Q. Traditional utilization, botany, phytochemistry, pharmacology, pharmaceutical analysis, processing and application of the seeds of Herpetospermum pedunculosum (Ser.) C.B. Clarke: a comprehensive review. Front Pharmacol 2024; 15:1498768. [PMID: 39749198 PMCID: PMC11694147 DOI: 10.3389/fphar.2024.1498768] [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: 09/19/2024] [Accepted: 11/26/2024] [Indexed: 01/04/2025] Open
Abstract
The seed of Herpetospermum pedunculosum (Ser.) C.B. Clarke, known in Chinese as Bo-Leng-Gua-Zi and in Tibetan as Sejimedo, are here abbreviated as H. pedunculosum seeds. Herpetospermum pedunculosum seeds is a traditional Chinese medicine for protecting the liver, clearing heat, and detoxifying. A total of 125 chemical metabolites of H. pedunculosum seeds are found, including lignans, fatty acids, terpenes, coumarins, and others. The pharmacological activities of H. pedunculosum seeds are mainly in hepatoprotective, antioxidant, anti-cancer cells, and anticholestatic effects. In clinical application, it is mainly used in combination with other traditional Chinese medicines to play a key role in treating the liver disease. This paper gives a systematic review of above research aspects, proposes the potential limitations and put forward plausible solutions. Relevant literatures were searched in PubMed, Web of Science and Chinese National Knowledge Infrastructure with Herpetospermum as the key word. A number of studies have shown that H. pedunculosum seeds exert excellent hepatoprotective effects by acting on NF-κB, TGF-β, and Keap1-Nrf2 signaling pathways, which provide a solid base for its clinic application. However, more research is needed to explore the standard cultivation and quality evaluation of H. pedunculosum seeds and systematical structure-activity relationship of its active metabolites.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Xiongyu Meng
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lupin Qin
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qiming Zhao
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
9
|
Shen J, Xie E, Shen S, Song Z, Li X, Wang F, Min J. Essentiality of SLC7A11-mediated nonessential amino acids in MASLD. Sci Bull (Beijing) 2024; 69:3700-3716. [PMID: 39366830 DOI: 10.1016/j.scib.2024.09.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 06/27/2024] [Accepted: 09/13/2024] [Indexed: 10/06/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) remains a rapidly growing global health burden. Here, we report that the nonessential amino acid (NEAA) transporter SLC7A11 plays a key role in MASLD. In patients with MASLD, we found high expression levels of SLC7A11 that were correlated directly with clinical grade. Using both loss-of-function and gain-of-function genetic models, we found that Slc7a11 deficiency accelerated MASLD progression via classic cystine/cysteine deficiency-induced ferroptosis, while serine deficiency and a resulting impairment in de novo cysteine production were attributed to ferroptosis-induced MASLD progression in mice overexpressing hepatic Slc7a11. Consistent with these findings, we found that both serine supplementation and blocking ferroptosis significantly alleviated MASLD, and the serum serine/glutamate ratio was significantly lower in these preclinical disease models, suggesting that it might serve as a prognostic biomarker for MASLD in patients. These findings indicate that defects in NEAA metabolism are involved in the progression of MASLD and that serine deficiency-triggered ferroptosis may provide a therapeutic target for its treatment.
Collapse
Affiliation(s)
- Jie Shen
- The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Enjun Xie
- The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Zhejiang University School of Medicine, Hangzhou 310058, China; The Second Affiliated Hospital, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou 310058, China; School of Public Health, School of Basic Medical Sciences, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, China; School of Public Health, School of Basic Medical Sciences, The First Affiliated Hospital, Xinxiang Medical University, Xinxiang 453003, China
| | - Shuying Shen
- The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Zijun Song
- The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xiaopeng Li
- The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Fudi Wang
- The Second Affiliated Hospital, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou 310058, China; School of Public Health, School of Basic Medical Sciences, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, China; School of Public Health, School of Basic Medical Sciences, The First Affiliated Hospital, Xinxiang Medical University, Xinxiang 453003, China.
| | - Junxia Min
- The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Zhejiang University School of Medicine, Hangzhou 310058, China.
| |
Collapse
|
10
|
Jian H, Li R, Huang X, Li J, Li Y, Ma J, Zhu M, Dong X, Yang H, Zou X. Branched-chain amino acids alleviate NAFLD via inhibiting de novo lipogenesis and activating fatty acid β-oxidation in laying hens. Redox Biol 2024; 77:103385. [PMID: 39426289 PMCID: PMC11536022 DOI: 10.1016/j.redox.2024.103385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/02/2024] [Accepted: 10/02/2024] [Indexed: 10/21/2024] Open
Abstract
The adverse metabolic impacts of branched-chain amino acids (BCAA) have been elucidated are mediated by isoleucine and valine. Dietary restriction of isoleucine promotes metabolic health and increases lifespan. However, a high protein diet enriched in BCAA is presently the most useful therapeutic strategy for nonalcoholic fatty liver disease (NAFLD), yet, its underlying mechanism remains largely unknown. Fatty liver hemorrhagic syndrome (FLHS), a specialized laying hen NAFLD model, can spontaneously develop fatty liver and hepatic steatosis under a high-energy and high-protein dietary background that the pathogenesis of FLHS is similar to human NAFLD. The mechanism underlying dietary BCAA control of NAFLD development in laying hens remains unclear. Herein, we demonstrate that dietary supplementation with 67 % High BCAA has unique mitigative impacts on NAFLD in laying hens. A High BCAA diet alleviates NAFLD, by inhibiting the tryptophan-ILA-AHR axis and MAPK9-mediated de novo lipogenesis (DNL), promoting ketogenesis and energy metabolism, and activating PPAR-RXR and pexophagy to promote fatty acid β-oxidation. Furthermore, we uncover that High BCAA strongly activates ubiquitin-proteasome autophagy via downregulating UFMylation to trigger MAPK9-mediated DNL, fatty acid elongation and lipid droplet formation-related proteins ubiquitination degradation, activating PPAR-RXR and pexophagy mediated fatty acid β-oxidation and lipolysis. Together, our data highlight moderating intake of high BCAA by inhibiting the AHR/MAPK9 are promising new strategies in NAFLD and FLHS treatment.
Collapse
Affiliation(s)
- Huafeng Jian
- Institute of Feed Science, College of Animal Sciences, Zhejiang University, Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Hangzhou, 310058, China; Xianghu Laboratory, Hangzhou, 311231, China
| | - Ru Li
- Institute of Feed Science, College of Animal Sciences, Zhejiang University, Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Hangzhou, 310058, China
| | - Xuan Huang
- Institute of Feed Science, College of Animal Sciences, Zhejiang University, Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Hangzhou, 310058, China
| | - Jiankui Li
- Institute of Feed Science, College of Animal Sciences, Zhejiang University, Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Hangzhou, 310058, China
| | - Yan Li
- Institute of Feed Science, College of Animal Sciences, Zhejiang University, Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Hangzhou, 310058, China
| | | | - Mingkun Zhu
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, 212100, China; Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, The Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, 212100, China
| | - Xinyang Dong
- Institute of Feed Science, College of Animal Sciences, Zhejiang University, Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Hangzhou, 310058, China
| | - Hua Yang
- Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, China.
| | - Xiaoting Zou
- Institute of Feed Science, College of Animal Sciences, Zhejiang University, Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Hangzhou, 310058, China.
| |
Collapse
|
11
|
Yang Y, Wu J, Zhou W, Ji G, Dang Y. Protein posttranslational modifications in metabolic diseases: basic concepts and targeted therapies. MedComm (Beijing) 2024; 5:e752. [PMID: 39355507 PMCID: PMC11442990 DOI: 10.1002/mco2.752] [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: 05/25/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 10/03/2024] Open
Abstract
Metabolism-related diseases, including diabetes mellitus, obesity, hyperlipidemia, and nonalcoholic fatty liver disease, are becoming increasingly prevalent, thereby posing significant threats to human health and longevity. Proteins, as the primary mediators of biological activities, undergo various posttranslational modifications (PTMs), including phosphorylation, ubiquitination, acetylation, methylation, and SUMOylation, among others, which substantially diversify their functions. These modifications are crucial in the physiological and pathological processes associated with metabolic disorders. Despite advancements in the field, there remains a deficiency in contemporary summaries addressing how these modifications influence processes of metabolic disease. This review aims to systematically elucidate the mechanisms through which PTM of proteins impact the progression of metabolic diseases, including diabetes, obesity, hyperlipidemia, and nonalcoholic fatty liver disease. Additionally, the limitations of the current body of research are critically assessed. Leveraging PTMs of proteins provides novel insights and therapeutic targets for the prevention and treatment of metabolic disorders. Numerous drugs designed to target these modifications are currently in preclinical or clinical trials. This review also provides a comprehensive summary. By elucidating the intricate interplay between PTMs and metabolic pathways, this study advances understanding of the molecular mechanisms underlying metabolic dysfunction, thereby facilitating the development of more precise and effective disease management strategies.
Collapse
Affiliation(s)
- Yunuo Yang
- Institute of Digestive DiseasesChina‐Canada Center of Research for Digestive Diseases (ccCRDD)Shanghai University of Traditional Chinese MedicineShanghaiChina
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine)ShanghaiChina
| | - Jiaxuan Wu
- Institute of Digestive DiseasesChina‐Canada Center of Research for Digestive Diseases (ccCRDD)Shanghai University of Traditional Chinese MedicineShanghaiChina
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine)ShanghaiChina
| | - Wenjun Zhou
- Institute of Digestive DiseasesChina‐Canada Center of Research for Digestive Diseases (ccCRDD)Shanghai University of Traditional Chinese MedicineShanghaiChina
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine)ShanghaiChina
| | - Guang Ji
- Institute of Digestive DiseasesChina‐Canada Center of Research for Digestive Diseases (ccCRDD)Shanghai University of Traditional Chinese MedicineShanghaiChina
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine)ShanghaiChina
| | - Yanqi Dang
- Institute of Digestive DiseasesChina‐Canada Center of Research for Digestive Diseases (ccCRDD)Shanghai University of Traditional Chinese MedicineShanghaiChina
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine)ShanghaiChina
| |
Collapse
|
12
|
Wang Y, Zhou J, Yang Q, Li X, Qiu Y, Zhang Y, Liu M, Zhu AJ. Therapeutic siRNA targeting PLIN2 ameliorates steatosis, inflammation, and fibrosis in steatotic liver disease models. J Lipid Res 2024; 65:100635. [PMID: 39187042 PMCID: PMC11440260 DOI: 10.1016/j.jlr.2024.100635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 07/10/2024] [Accepted: 08/13/2024] [Indexed: 08/28/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most prevalent chronic liver disease worldwide. If left untreated, MASLD can progress from simple hepatic steatosis to metabolic dysfunction-associated steatohepatitis, which is characterized by inflammation and fibrosis. Current treatment options for MASLD remain limited, leaving substantial unmet medical needs for innovative therapeutic approaches. Here, we show that PLIN2, a lipid droplet protein inhibiting hepatic lipolysis, serves as a promising therapeutic target for MASLD. Hepatic PLIN2 levels were markedly elevated in multiple MASLD mouse models induced by diverse nutritional and genetic factors. The liver-specific deletion of Plin2 exhibited significant anti-MASLD effects in these models. To translate this discovery into a therapeutic application, we developed a GalNAc-siRNA conjugate with enhanced stabilization chemistry and validated its potent and sustained efficacy in suppressing Plin2 expression in mouse livers. This siRNA therapeutic, named GalNAc-siPlin2, was shown to be biosafe in mice. Treatment with GalNAc-siPlin2 for 6-8 weeks led to a decrease in hepatic triglyceride levels by approximately 60% in high-fat diet- and obesity-induced MASLD mouse models, accompanied with increased hepatic secretion of VLDL-triglyceride and enhanced thermogenesis in brown adipose tissues. Eight-week treatment with GalNAc-siPlin2 significantly improved hepatic steatosis, inflammation, and fibrosis in high-fat/high fructose-induced metabolic dysfunction-associated steatohepatitis models compared to control group. As a proof of concept, we developed a GalNAc-siRNA therapeutic targeting human PLIN2, which effectively suppressed hepatic PLIN2 expression and ameliorated MASLD in humanized PLIN2 knockin mice. Together, our results highlight the potential of GalNAc-siPLIN2 as a candidate MASLD therapeutic for clinical trials.
Collapse
Affiliation(s)
- Yao Wang
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, China
| | - Jiaxin Zhou
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, China
| | - Qi Yang
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, China
| | - Xinmeng Li
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Yifu Qiu
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China; Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Yansong Zhang
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, China; Peking University Chengdu Academy for Advanced Interdisciplinary Biotechnologies, Chengdu, Sichuan, China.
| | - Min Liu
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China.
| | - Alan Jian Zhu
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China; Peking University Chengdu Academy for Advanced Interdisciplinary Biotechnologies, Chengdu, Sichuan, China.
| |
Collapse
|
13
|
Wang Y, Gu W, Xu Z, Lv L, Wang D, Jin Y, Wang X. Comprehensive multi-omics investigation of sub-chronic toxicity induced by Cadmium and Triazophos Co-exposure in hook snout carps (Opsariichthys bidens). JOURNAL OF HAZARDOUS MATERIALS 2024; 476:135104. [PMID: 38970972 DOI: 10.1016/j.jhazmat.2024.135104] [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: 05/07/2024] [Revised: 06/18/2024] [Accepted: 07/03/2024] [Indexed: 07/08/2024]
Abstract
The coexistence of heavy metals and pesticides poses a critical challenge in agricultural ecosystems. Traditional toxicity assessments often focus only on the individual impacts of either pesticides or heavy metals. Here, the untargeted metabolomics and 16 S rRNA sequencing were used to assess the individual and combined effects of cadmium (Cd) and triazophos (TRI) on hook snout carps (Opsariichthys bidens). Cd caused much more serious impacts on hepatic metabolism and gut microbiota than those in TRI. Combined Cd and TRI exposure synergistically affected hepatic metabolism, causing mitochondrial dysfunction and even oxidative damage. Simultaneously, 16 S rRNA sequencing highlighted significant variations in the composition and abundance of gut microbiota. A noteworthy connection emerged between these distinct microbiota profiles and disruptions in energy metabolism, ultimately leading to disorders in metabolites. These findings enhanced the understanding of risks posed by heavy metals and pesticides, providing insights for better environmental risk assessments of aquatic organisms.
Collapse
Affiliation(s)
- Yanhua Wang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Quality and Standard for Agro-products, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, Zhejiang, PR China
| | - Weijie Gu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, Zhejiang, PR China
| | - Zhenlan Xu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Quality and Standard for Agro-products, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, Zhejiang, PR China
| | - Lu Lv
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Quality and Standard for Agro-products, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, Zhejiang, PR China
| | - Dou Wang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Quality and Standard for Agro-products, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, Zhejiang, PR China
| | - Yuanxiang Jin
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, Zhejiang, PR China.
| | - Xinquan Wang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Quality and Standard for Agro-products, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, Zhejiang, PR China.
| |
Collapse
|
14
|
Yuan W, Han J, Chen C, Qiu Y, Xu Y, Huang Y, Chen Z, Xu A, Sun M. UBR1 is a prognostic biomarker and therapeutic target associated with immune cell infiltration in gastric cancer. Aging (Albany NY) 2024; 16:12029-12049. [PMID: 39181686 PMCID: PMC11386912 DOI: 10.18632/aging.206079] [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: 12/19/2023] [Accepted: 07/15/2024] [Indexed: 08/27/2024]
Abstract
BACKGROUND Ubiquitination is a targeted protein modification process mediated by intracellular molecules. UBR1 encodes a protein that binds to unstable N-terminal residues of substrate proteins and contributes to the formation of substrate-linked polyubiquitin chains. However, the function and cellular pathways of UBR1 in tumors have received inadequate attention. This study aimed to investigate the potential of UBR1 as a prognostic biomarker and immunotherapy target for stomach adenocarcinoma (STAD) as well as its biological function and molecular mechanism in relation to the disease. METHODS Differential expression and pan-cancer gene set enrichment analysis (GSEA) were conducted using The Cancer Genome Atlas (TCGA), Gene Expression Omnibus (GEO), and Genotype-Tissue Expression (GTEx) datasets. The Human Protein Atlas (HPA) database was utilized to identify UBR1-enriched pathways in AGS cells and to compare immunohistochemical differences between cancerous and adjacent non-cancerous tissues in gastric cancer. Quantitative Polymerase Chain Reaction (QPCR) and Western blot (WB) analyses were employed to validate these findings in both cancerous and adjacent non-cancerous tissues of gastric cancer. UBR1 expression in GES-1 and four gastric cancer cell lines was assessed using QPCR and WB. Kaplan-Meier curves, univariate and multivariate Cox regression analyses, and receiver operating characteristic (ROC) curve analyses were performed to evaluate the prognostic and diagnostic roles of UBR1. Additionally, the correlation between UBR1 expression and clinical parameters was analyzed using TCGA and GEO databases. UBR1 mutation data were obtained from the cBioPortal database. The mutation landscape, mutation-associated genes, protein structure, tumor mutation burden (TMB), and microsatellite instability (MSI) correlations were analyzed and illustrated. The biological functions of UBR1 were investigated using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses. The correlation between UBR1 and immune infiltration was assessed using TIMER and EPIC computational methods. Protein expression levels of UBR1 in gastric cancer cell lines were determined by immunohistochemistry (IHC) and WB analysis. Quantitative real-time PCR (qRT-PCR) was employed to analyze mRNA expression. Immunoprecipitation (IP) assays were conducted to detect protein-protein interactions between UBR1 and PDL1, while cellular immunofluorescence was used to observe the co-localization of these proteins. Cell proliferation was evaluated using CCK8 and colony formation assays. Cell migration was assessed using Transwell and wound healing assays. Finally, apoptosis was analyzed using flow cytometry, and WB was used to detect changes in apoptotic proteins and NF-κB P65 pathway proteins. RESULTS UBR1 was upregulated in 28 cancer types, including STAD, and its overexpression was validated in gastric cancer cell lines and tissues. UBR1 expression was associated with advanced pathological characteristics. High UBR1 expression was linked to poor prognostic outcomes, including overall survival (OS), progression-free interval (PFI), disease-specific survival (DSS), as well as responses to surgery, chemotherapy, and HER2 expression. UBR1 expression showed significant correlations with clinical parameters such as age, gender, TNM stage, pathological stage, tumor resection, and anti-reflux therapy. Amplifications and deletions were the most frequent genetic alterations associated with UBR1. According to KEGG and GSEA analyses, UBR1 was significantly associated with several cancer pathways, oxidative phosphorylation, and the TNF-NFκB pathway. UBR1 also exhibited a significant correlation with immune cell infiltration and immunotherapy, including a direct interaction with PDL1. Knockdown of UBR1 inhibited the proliferation, migration, and invasion of STAD cells and promoted apoptosis. CONCLUSIONS UBR1 is overexpressed in STAD, promoting its progression and positively correlating with immune cell infiltration and immunotherapeutic responses. Therefore, UBR1 could be a promising biomarker for the prognosis and immunotherapy of STAD.
Collapse
Affiliation(s)
- Weiwei Yuan
- Department of Thyroid Surgery, Baoshan Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201999, China
| | - Jianye Han
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Chen Chen
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Anhui Public Health Clinical Center, Hefei 230012, China
| | - Yue Qiu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Yuanmin Xu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Yang Huang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Anhui Public Health Clinical Center, Hefei 230012, China
| | - Zhangming Chen
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Aman Xu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Minzhi Sun
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Anhui Public Health Clinical Center, Hefei 230012, China
| |
Collapse
|
15
|
Zhu X, Lin M, Chi Y, Li X, Jiang Z, Jian X, Lian M, Wu X, Han S, Shi X. Qualitative and quantitative analysis of chemical components in Qianggan capsule by UHPLC-Q-TOF-MS/MS and LC-sMRM. J Chromatogr A 2024; 1728:465020. [PMID: 38805896 DOI: 10.1016/j.chroma.2024.465020] [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: 02/11/2024] [Revised: 05/08/2024] [Accepted: 05/22/2024] [Indexed: 05/30/2024]
Abstract
Qianggan capsule (QGC) is a complex preparation composed of 16 traditional Chinese medicines (TCM) that can clear heat and dampness, fortify the spleen and blood, typify qi and relieve depression. However, the chemical composition of QGC remains incompletely understood, despite its clinical use in treating chronic hepatitis and liver injury. The objective of this study was to explore the quality markers of QGC through qualitative and quantitative analysis of its chemical components. First, the chemical composition of QGC was qualitatively analyzed using UHPLC-Q-TOF-MS/MS. Subsequently, the LC-sMRM method was developed and optimized to accurately quantify various chemical components of 10 batches of QGC. Finally, the variations in chemical components between batches were analyzed via multivariate statistical analysis. UHPLC-Q-TOF-MS/MS analysis revealed 167 chemical constituents in QGC, comprised of 48 flavonoids, 32 terpenoids, 18 phenolic acids, 9 coumarins, 9 phenylpropanoids, and 51 nucleosides, sugars, amino acids, anthraquinones, and other compounds. The LC-sMRM method was established for the quantitative analysis of 42 chemical components in 10 batches of QGC. The ultrasonic-assisted extraction parameters were optimized using RSM. Compared with conventional MRM, sMRM demonstrated superior sensitivity and precision. PCA and OPLS-DA identified eight chemical components with content differences among batches. This study established the chemical composition of QGC, offering useful guidance for assessing its quality.
Collapse
Affiliation(s)
- Xiaoliang Zhu
- Hebei Key Laboratory of Innovative Drug Development and Evaluation, School of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang 050017, China; National Demonstration Center for Experimental Pharmacy Education, School of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang 050017, China
| | - Mengmeng Lin
- Hebei Key Laboratory of Innovative Drug Development and Evaluation, School of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang 050017, China; National Demonstration Center for Experimental Pharmacy Education, School of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang 050017, China
| | - Yuqian Chi
- Hebei Key Laboratory of Innovative Drug Development and Evaluation, School of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang 050017, China; National Demonstration Center for Experimental Pharmacy Education, School of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang 050017, China
| | - Xin Li
- Hebei Key Laboratory of Innovative Drug Development and Evaluation, School of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang 050017, China; National Demonstration Center for Experimental Pharmacy Education, School of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang 050017, China
| | - Ziyi Jiang
- Hebei Key Laboratory of Innovative Drug Development and Evaluation, School of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang 050017, China; National Demonstration Center for Experimental Pharmacy Education, School of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang 050017, China
| | - Xiaoyang Jian
- Hebei Key Laboratory of Innovative Drug Development and Evaluation, School of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang 050017, China; National Demonstration Center for Experimental Pharmacy Education, School of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang 050017, China
| | - Mengyuan Lian
- Hebei Key Laboratory of Innovative Drug Development and Evaluation, School of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang 050017, China; National Demonstration Center for Experimental Pharmacy Education, School of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang 050017, China
| | - Xiaodi Wu
- Hebei Key Laboratory of Innovative Drug Development and Evaluation, School of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang 050017, China; National Demonstration Center for Experimental Pharmacy Education, School of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang 050017, China
| | - Shuang Han
- Department of Teaching Support, Hebei Open University, Shijiazhuang 052360, China.
| | - Xiaowei Shi
- Hebei Key Laboratory of Innovative Drug Development and Evaluation, School of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang 050017, China; National Demonstration Center for Experimental Pharmacy Education, School of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang 050017, China; National Key Laboratory of New Pharmaceutical Preparations and excipients, School of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang 050017, China.
| |
Collapse
|
16
|
Li M, Zhang J, Li Z, Xu Z, Qian S, Tay LJ, Zhang Z, Yang F, Huang Y. The role and mechanism of SUMO modification in liver disease. Biomed Pharmacother 2024; 177:116898. [PMID: 38878635 DOI: 10.1016/j.biopha.2024.116898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 06/04/2024] [Accepted: 06/06/2024] [Indexed: 07/28/2024] Open
Abstract
Liver disease affects millions of people in the world, and China has the highest prevalence of liver disease in the world. Small ubiquitin-related modifier (SUMO) modification is a highly conserved post-translational modification of proteins. They are widely expressed in a variety of tissues, including the heart, liver, kidney and lung. SUMOylation of protein plays a key role in the occurrence and development of liver disease. Therefore, this study reviewed the effects of SUMO protein on non-alcoholic fatty liver disease (NAFLD), alcoholic liver disease (ALD), viral hepatitis, hepatic fibrosis (HF), hepatocellular carcinoma (HCC), and other liver diseases to provide novel strategies for targeted treatment of liver disease.
Collapse
Affiliation(s)
- Mengxue Li
- Anhui Provincial laboratory of inflammatory and immunity Disease, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China
| | - Jingrong Zhang
- Anhui Provincial laboratory of inflammatory and immunity Disease, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China
| | - Zihao Li
- Anhui Provincial laboratory of inflammatory and immunity Disease, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China
| | - Zhou Xu
- Anhui Provincial laboratory of inflammatory and immunity Disease, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China
| | - Shishun Qian
- Anhui Provincial laboratory of inflammatory and immunity Disease, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China
| | - Lynn Jia Tay
- School of International Education, Anhui Medical University, Hefei 230032, China
| | - Ziwen Zhang
- Anhui Provincial laboratory of inflammatory and immunity Disease, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China
| | - Furong Yang
- Anhui Provincial laboratory of inflammatory and immunity Disease, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China.
| | - Yan Huang
- Anhui Provincial laboratory of inflammatory and immunity Disease, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China; School of International Education, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
17
|
Yan L, Yan Y, Yang K, Chang Q, Zhang L. Metabolomics reveals dysregulated all-trans retinoic acid and polyunsaturated fatty acid metabolism contribute to PXR-induced hepatic steatosis in mice. Toxicol Lett 2024; 398:150-160. [PMID: 38971454 DOI: 10.1016/j.toxlet.2024.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 06/05/2024] [Accepted: 07/02/2024] [Indexed: 07/08/2024]
Abstract
Activation of pregnane X receptor (PXR) by xenobiotics has been associated with metabolic diseases. This study aimed to reveal the impact of PXR activation on hepatic metabolome and explore novel mechanisms underlying PXR-mediated lipid metabolism disorder in the liver. Wild-type and PXR-deficient male C57BL/6 mice were used as in vivo models, and hepatic steatosis was induced by pregnenolone-16α-carbonitrile, a typical rodent PXR agonist. Metabolomic analysis of liver tissues showed that PXR activation led to significant changes in metabolites involved in multiple metabolic pathways previously reported, including lipid metabolism, energy homeostasis, and amino acid metabolism. Moreover, the level of hepatic all-trans retinoic acid (ATRA), the main active metabolite of vitamin A, was significantly increased by PXR activation, and genes involved in ATRA metabolism exhibited differential expression following PXR activation or deficiency. Consistent with previous research, the expression of downstream target genes of peroxisome proliferator-activated receptor α (PPARα) was decreased. Analysis of fatty acids by Gas Chromatography-Mass Spectrometer further revealed changes in polyunsaturated fatty acid metabolism upon PXR activation, suggesting inhibition of PPARα activity. Taken together, our findings reveal a novel metabolomic signature of hepatic steatosis induced by PXR activation in mice.
Collapse
Affiliation(s)
- Liang Yan
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou Univerisity, Zhengzhou 450052, China.
| | - Yachun Yan
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou Univerisity, Zhengzhou 450052, China
| | - Kun Yang
- Department of Pharmacology, School of Basic Medicine, Zhengzhou University, Zhengzhou 450052, China
| | - Qi Chang
- Department of Pharmacology, School of Basic Medicine, Zhengzhou University, Zhengzhou 450052, China
| | - Lirong Zhang
- Department of Pharmacology, School of Basic Medicine, Zhengzhou University, Zhengzhou 450052, China.
| |
Collapse
|
18
|
Govatati S, Kumar R, Boro M, Traylor JG, Orr AW, Lusis AJ, Rao GN. TRIM13 reduces cholesterol efflux and increases oxidized LDL uptake leading to foam cell formation and atherosclerosis. J Biol Chem 2024; 300:107224. [PMID: 38537695 PMCID: PMC11053335 DOI: 10.1016/j.jbc.2024.107224] [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: 02/14/2024] [Revised: 03/11/2024] [Accepted: 03/13/2024] [Indexed: 04/25/2024] Open
Abstract
Impaired cholesterol efflux and/or uptake can influence arterial lipid accumulation leading to atherosclerosis. Here, we report that tripartite motif-containing protein 13 (TRIM13), a RING-type E3 ubiquitin ligase, plays a role in arterial lipid accumulation leading to atherosclerosis. Using molecular approaches and KO mouse model, we found that TRIM13 expression was induced both in the aorta and peritoneal macrophages (pMφ) of ApoE-/- mice in response to Western diet (WD) in vivo. Furthermore, proatherogenic cytokine interleukin-1β also induced TRIM13 expression both in pMφ and vascular smooth muscle cells. Furthermore, we found that TRIM13 via ubiquitination and degradation of liver X receptor (LXR)α/β downregulates the expression of their target genes ABCA1/G1 and thereby inhibits cholesterol efflux. In addition, TRIM13 by ubiquitinating and degrading suppressor of cytokine signaling 1/3 (SOCS1/3) mediates signal transducer and activator of transcription 1 (STAT1) activation, CD36 expression, and foam cell formation. In line with these observations, genetic deletion of TRIM13 by rescuing cholesterol efflux and inhibiting foam cell formation protects against diet-induced atherosclerosis. We also found that while TRIM13 and CD36 levels were increased, LXRα/β, ABCA1/G1, and SOCS3 levels were decreased both in Mφ and smooth muscle cells of stenotic human coronary arteries as compared to nonstenotic arteries. More intriguingly, the expression levels of TRIM13 and its downstream signaling molecules were correlated with the severity of stenotic lesions. Together, these observations reveal for the first time that TRIM13 plays a crucial role in diet-induced atherosclerosis, and that it could be a potential drug target against this vascular lesion.
Collapse
Affiliation(s)
- Suresh Govatati
- Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Raj Kumar
- Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Monoranjan Boro
- Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - James G Traylor
- Department of Pathology, Louisiana State University Health Science Center, Shreveport, Louisiana, USA
| | - A Wayne Orr
- Department of Pathology, Louisiana State University Health Science Center, Shreveport, Louisiana, USA
| | - Aldons J Lusis
- Division of Cardiology, Department of Medicine, University of California, Los Angeles, California, USA
| | - Gadiparthi N Rao
- Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee, USA.
| |
Collapse
|
19
|
Tyczyńska M, Hunek G, Szczasny M, Brachet A, Januszewski J, Forma A, Portincasa P, Flieger J, Baj J. Supplementation of Micro- and Macronutrients-A Role of Nutritional Status in Non-Alcoholic Fatty Liver Disease. Int J Mol Sci 2024; 25:4916. [PMID: 38732128 PMCID: PMC11085010 DOI: 10.3390/ijms25094916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 04/28/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a condition in which the pathological cumulation of fat with coexisting inflammation and damage of hepatic cells leads to progressive dysfunctions of the liver. Except for the commonly well-known major causes of NAFLD such as obesity, dyslipidemia, insulin resistance, or diabetes, an unbalanced diet and imbalanced nutritional status should also be taken into consideration. In this narrative review, we summarized the current knowledge regarding the micro- and macronutrient status of patients suffering from NAFLD considering various diets and supplementation of chosen supplements. We aimed to summarize the knowledge indicating which nutritional impairments may be associated with the onset and progression of NAFLD at the same time evaluating the potential therapy targets that could facilitate the healing process. Except for the above-mentioned objectives, one of the most important aspects of this review was to highlight the possible strategies for taking care of NAFLD patients taking into account the challenges and opportunities associated with the micronutrient status of the patients. The current research indicates that a supplementation of chosen vitamins (e.g., vitamin A, B complex, C, or D) as well as chosen elements such as zinc may alleviate the symptoms of NAFLD. However, there is still a lack of sufficient data regarding healthy ranges of dosages; thus, further research is of high importance in this matter.
Collapse
Affiliation(s)
- Magdalena Tyczyńska
- Department of Correct, Clinical and Imaging Anatomy, Medical University of Lublin, Jaczewskiego 4, 20-090 Lublin, Poland;
| | - Gabriela Hunek
- Chair and Department of Forensic Medicine, Medical University of Lublin, Jaczewskiego 8b, 20-090 Lublin, Poland; (G.H.); (A.B.)
| | - Martyna Szczasny
- Chair and Department of Anatomy, Medical University of Lublin, Jaczewskiego 4, 20-090 Lublin, Poland; (M.S.); (J.J.)
| | - Adam Brachet
- Chair and Department of Forensic Medicine, Medical University of Lublin, Jaczewskiego 8b, 20-090 Lublin, Poland; (G.H.); (A.B.)
| | - Jacek Januszewski
- Chair and Department of Anatomy, Medical University of Lublin, Jaczewskiego 4, 20-090 Lublin, Poland; (M.S.); (J.J.)
| | - Alicja Forma
- Chair and Department of Forensic Medicine, Medical University of Lublin, Jaczewskiego 8b, 20-090 Lublin, Poland; (G.H.); (A.B.)
| | - Piero Portincasa
- Clinica Medica “A. Murri”, Department of Biomedical Sciences & Human Oncology, University of Bari Medical School, 70124 Bari, Italy;
| | - Jolanta Flieger
- Department of Analytical Chemistry, Medical University of Lublin, Chodźki 4A, 20-093 Lublin, Poland;
| | - Jacek Baj
- Chair and Department of Anatomy, Medical University of Lublin, Jaczewskiego 4, 20-090 Lublin, Poland; (M.S.); (J.J.)
| |
Collapse
|
20
|
Gao J, Zhang S, Deng P, Wu Z, Lemaitre B, Zhai Z, Guo Z. Dietary L-Glu sensing by enteroendocrine cells adjusts food intake via modulating gut PYY/NPF secretion. Nat Commun 2024; 15:3514. [PMID: 38664401 PMCID: PMC11045819 DOI: 10.1038/s41467-024-47465-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 03/28/2024] [Indexed: 04/28/2024] Open
Abstract
Amino acid availability is monitored by animals to adapt to their nutritional environment. Beyond gustatory receptors and systemic amino acid sensors, enteroendocrine cells (EECs) are believed to directly percept dietary amino acids and secrete regulatory peptides. However, the cellular machinery underlying amino acid-sensing by EECs and how EEC-derived hormones modulate feeding behavior remain elusive. Here, by developing tools to specifically manipulate EECs, we find that Drosophila neuropeptide F (NPF) from mated female EECs inhibits feeding, similar to human PYY. Mechanistically, dietary L-Glutamate acts through the metabotropic glutamate receptor mGluR to decelerate calcium oscillations in EECs, thereby causing reduced NPF secretion via dense-core vesicles. Furthermore, two dopaminergic enteric neurons expressing NPFR perceive EEC-derived NPF and relay an anorexigenic signal to the brain. Thus, our findings provide mechanistic insights into how EECs assess food quality and identify a conserved mode of action that explains how gut NPF/PYY modulates food intake.
Collapse
Affiliation(s)
- Junjun Gao
- Department of Medical Genetics, School of Basic Medicine, Institute for Brain Research, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Song Zhang
- Department of Medical Genetics, School of Basic Medicine, Institute for Brain Research, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pan Deng
- State Key Laboratory of Digital Manufacturing Equipment and Technology, Huazhong University of Science and Technology, Wuhan, PR China
- Department of Mechanical Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| | - Zhigang Wu
- State Key Laboratory of Digital Manufacturing Equipment and Technology, Huazhong University of Science and Technology, Wuhan, PR China
| | - Bruno Lemaitre
- Global Health Institute, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Zongzhao Zhai
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, Hunan, PR China.
| | - Zheng Guo
- Department of Medical Genetics, School of Basic Medicine, Institute for Brain Research, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Cell Architecture Research Center, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
21
|
Yang K, Liu C, Shao J, Guo L, Wang Q, Meng Z, Jin X, Chen X. Would Combination Be Better: Swimming Exercise and Intermittent Fasting Improve High-Fat Diet-Induced Nonalcoholic Fatty Liver Disease in Obese Rats via the miR-122-5p/SREBP-1c/CPT1A Pathway. Diabetes Metab Syndr Obes 2024; 17:1675-1686. [PMID: 38623310 PMCID: PMC11016699 DOI: 10.2147/dmso.s448165] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 03/12/2024] [Indexed: 04/17/2024] Open
Abstract
Background Swimming and intermittent fasting can both improve obesity-induced NAFLD, but which of the two is more effective and whether the combination of the two has a superimposed effect is inconclusive. Methods The model of NAFLD in obese rats was established by a high-fat diet and performed swimming, intermittent fasting, and a combination of both interventions for 8 weeks. Serum lipids and enzyme activity were measured by an automatic biochemical analyzer. Liver morphostructural analysis was observed by transmission electron microscopy, and morphology was observed by HE staining. RT‒PCR was used to detect the mRNA level. Results Morphology and microstructure of the liver of model rats were impaired, with the upregulation of miR-122-5p, SREBP-1c, FASN and ACC1. Eight weeks of swimming exercise, intermittent fasting and the combination of both attenuate these effects, manifested by the downregulation of miR-122-5p and upregulation of CPT1A mRNA levels. There was no significant stacking effect of the combination of the swimming and intermittent fasting interventions. Conclusion NAFLD leads to pathology in model rats. Eight weeks of swimming exercise, intermittent fasting and the combination of both can inhibit miR-122-5p and improve hepatic lipid metabolism, while no significant additive effects of combining the interventions were found.
Collapse
Affiliation(s)
- Kang Yang
- Rehabilitation Medicine Department, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou city, Jiangsu Province, People’s Republic of China
| | - Chengye Liu
- Rehabilitation Medicine Department, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou city, Jiangsu Province, People’s Republic of China
| | - Jun Shao
- Cardiovascular Disease Center, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou city, Jiangsu Province, People’s Republic of China
| | - Lingxiang Guo
- Cardiovascular Disease Center, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou city, Jiangsu Province, People’s Republic of China
| | - Qing Wang
- Respiratory Department, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou city, Jiangsu Province, People’s Republic of China
| | - Zhaoxiang Meng
- Rehabilitation Medicine Department, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou city, Jiangsu Province, People’s Republic of China
| | - Xing Jin
- Rehabilitation Medicine Department, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou city, Jiangsu Province, People’s Republic of China
| | - Xianghe Chen
- College of Physical Education, Yangzhou University, Yangzhou city, Jiangsu Province, People’s Republic of China
| |
Collapse
|
22
|
Li J, Zhang Q, Guan Y, Liao D, Chen H, Xiong H, Sheng Y, Chen X, Pang J. TRIB3 promotes the progression of renal cell carcinoma by upregulating the lipid droplet-associated protein PLIN2. Cell Death Dis 2024; 15:240. [PMID: 38561354 PMCID: PMC10985002 DOI: 10.1038/s41419-024-06627-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 03/18/2024] [Accepted: 03/20/2024] [Indexed: 04/04/2024]
Abstract
Abnormal lipid metabolism and lipid accumulation are characteristic hallmarks of renal cell carcinoma (RCC). While there is prior evidence closely linking such lipid accumulation within RCC cells and consequent tumorigenesis, the mechanisms underlying this process remain incompletely understood. In this study, a series of bioinformatics analyses were initially performed by screening RCC databases and gene sets, ultimately leading to the identification of TRIB3 as an oncogene that functions as a central regulator of lipid metabolism. TRIB3 overexpression was observed in both RCC patient tumor tissues and cell lines, and this upregulation was correlated with a worse RCC patient prognosis. When TRIB3 was knocked down, this resulted in a reduction in lipid accumulation and the consequent induction of endoplasmic reticulum (ER) stress-related apoptotic cell death. At the molecular level, interactions between TRIB3 and PLIN2 were found to abrogate TEB4-mediated PLIN2 ubiquitination and consequent degradation, thus maintaining higher PLIN2 expression levels. This simultaneously helps facilitate the accumulation of lipids while preserving ER homeostasis, thus driving accelerated RCC tumor progression. This TRIB3-PLIN2 axis thus represents a promising new target for efforts to treat RCC.
Collapse
Affiliation(s)
- Jun Li
- Department of Urology, Kidney and Urology Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China
| | - Qian Zhang
- Department of Rehabilitation Medicine, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China
| | - Yupeng Guan
- Department of Urology, Kidney and Urology Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China
| | - Dingzhun Liao
- Department of Pathology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China
| | - Huikun Chen
- Department of Urology, Kidney and Urology Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China
| | - Haiyun Xiong
- Department of Urology, Kidney and Urology Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China
| | - Yiyu Sheng
- Department of Urology, Kidney and Urology Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China
| | - Xianju Chen
- Department of Urology, Kidney and Urology Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China
| | - Jun Pang
- Department of Urology, Kidney and Urology Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China.
| |
Collapse
|
23
|
Zou Y, Zhang Y, Li M, Cao K, Song C, Zhang Z, Cai K, Geng D, Chen S, Wu Y, Zhang N, Sun G, Wang J, Zhang Y, Sun Y. Regulation of lipid metabolism by E3 ubiquitin ligases in lipid-associated metabolic diseases. Int J Biol Macromol 2024; 265:130961. [PMID: 38508558 DOI: 10.1016/j.ijbiomac.2024.130961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 03/10/2024] [Accepted: 03/15/2024] [Indexed: 03/22/2024]
Abstract
Previous studies have progressively elucidated the involvement of E3 ubiquitin (Ub) ligases in regulating lipid metabolism. Ubiquitination, facilitated by E3 Ub ligases, modifies critical enzymes in lipid metabolism, enabling them to respond to specific signals. In this review, we aim to present a comprehensive analysis of the role of E3 Ub ligases in lipid metabolism, which includes lipid synthesis and lipolysis, and their influence on cellular lipid homeostasis through the modulation of lipid uptake and efflux. Furthermore, it explores how the ubiquitination process governs the degradation or activation of pivotal enzymes, thereby regulating lipid metabolism at the transcriptional level. Perturbations in lipid metabolism have been implicated in various diseases, including hepatic lipid metabolism disorders, atherosclerosis, diabetes, and cancer. Therefore, this review focuses on the association between E3 Ub ligases and lipid metabolism in lipid-related diseases, highlighting enzymes critically involved in lipid synthesis and catabolism, transcriptional regulators, lipid uptake translocators, and transporters. Overall, this review aims to identify gaps in current knowledge, highlight areas requiring further research, offer potential targeted therapeutic approaches, and provide a comprehensive outlook on clinical conditions associated with lipid metabolic diseases.
Collapse
Affiliation(s)
- Yuanming Zou
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Ying Zhang
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China; Institute of Health Sciences, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, 110001, Liaoning Province, People's Republic of China.
| | - Mohan Li
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Kexin Cao
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Chunyu Song
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Zhaobo Zhang
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Kexin Cai
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Danxi Geng
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Shuxian Chen
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Yanjiao Wu
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Naijin Zhang
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China; Institute of Health Sciences, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, 110001, Liaoning Province, People's Republic of China; Key Laboratory of Reproductive and Genetic Medicine (China Medical University), National Health Commission, 77 Puhe Road, Shenbei New District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Guozhe Sun
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China.
| | - Jing Wang
- Department of Hematology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China.
| | - Yixiao Zhang
- Department of Urology Surgery, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning Province, People's Republic of China.
| | - Yingxian Sun
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China; Institute of Health Sciences, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, 110001, Liaoning Province, People's Republic of China; Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, 110001, Liaoning Province, People's Republic of China.
| |
Collapse
|
24
|
Deng Y, Hu M, Huang S, Fu N. Molecular mechanism and therapeutic significance of essential amino acids in metabolically associated fatty liver disease. J Nutr Biochem 2024; 126:109581. [PMID: 38219809 DOI: 10.1016/j.jnutbio.2024.109581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 01/01/2024] [Accepted: 01/06/2024] [Indexed: 01/16/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD), also known as metabolically associated fatty liver disease (MAFLD), is a systemic metabolic disease characterized by lipid accumulation in the liver, lipid toxicity, insulin resistance, intestinal dysbiosis, and inflammation that can progress from simple steatosis to nonalcoholic steatohepatitis (NASH) and even cirrhosis or cancer. It is the most prevalent illness threatening world health. Currently, there are almost no approved drug interventions for MAFLD, mainly dietary changes and exercise to control weight and regulate metabolic disorders. Meanwhile, the metabolic pathway involved in amino acid metabolism also influences the onset and development of MAFLD in the body, and most amino acid metabolism takes place in the liver. Essential amino acids are those amino acids that must be supplemented from outside the diet and that cannot be synthesized in the body or cannot be synthesized at a rate sufficient to meet the body's needs, including leucine, isoleucine, valine (collectively known as branched-chain amino acids), tryptophan, phenylalanine (which are aromatic amino acids), histidine, methionine, threonine and lysine. The metabolic balance of the body is closely linked to these essential amino acids, and essential amino acids are closely linked to the pathophysiological process of MAFLD. In this paper, we will focus on the metabolism of essential amino acids in the body and further explore the therapeutic strategies for MAFLD based on the studies conducted in recent years.
Collapse
Affiliation(s)
- Yuting Deng
- The Affiliated Nanhua Hospital, Department of Gastroenterology, Hunan Provincial Clinical Research Center of Metabolic Associated Fatty Liver Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, 421002, China
| | - Mengsi Hu
- The Affiliated Nanhua Hospital, Department of Gastroenterology, Hunan Provincial Clinical Research Center of Metabolic Associated Fatty Liver Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, 421002, China
| | - Shufang Huang
- The Affiliated Nanhua Hospital, Hunan Provincial Clinical Research Center of Metabolic Associated Fatty Liver Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, 421002, China.
| | - Nian Fu
- The Affiliated Nanhua Hospital, Department of Gastroenterology, Hunan Provincial Clinical Research Center of Metabolic Associated Fatty Liver Disease, Hengyang Medical School, University of South China, Hengyang, Hunan, 421002, China; The Affiliated Nanhua Hospital, Institute of Clinical Research, Hengyang Medical School, University of South China, Hengyang, Hunan, 421002, China.
| |
Collapse
|
25
|
Wu T, Chen X, Xu K, Dai C, Li X, Zhang YWQ, Li J, Gao M, Liu Y, Liu F, Zhang X, Wang B, Xia P, Li Z, Ma W, Yuan Y. LIM domain only 7 negatively controls nonalcoholic steatohepatitis in the setting of hyperlipidemia. Hepatology 2024; 79:149-166. [PMID: 37676481 PMCID: PMC10718224 DOI: 10.1097/hep.0000000000000585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 07/19/2023] [Indexed: 09/08/2023]
Abstract
BACKGROUND AND AIMS Hyperlipidemia has been extensively recognized as a high-risk factor for NASH; however, clinical susceptibility to NASH is highly heterogeneous. The key controller(s) of NASH susceptibility in patients with hyperlipidemia has not yet been elucidated. Here, we aimed to reveal the key regulators of NASH in patients with hyperlipidemia and to explore its role and underlying mechanisms. APPROACH AND RESULTS To identify the predominant suppressors of NASH in the setting of hyperlipidemia, we collected liver biopsy samples from patients with hyperlipidemia, with or without NASH, and performed RNA-sequencing analysis. Notably, decreased Lineage specific Interacting Motif domain only 7 (LMO7) expression robustly correlated with the occurrence and severity of NASH. Although overexpression of LMO7 effectively blocked hepatic lipid accumulation and inflammation, LMO7 deficiency in hepatocytes greatly exacerbated diet-induced NASH progression. Mechanistically, lysine 48 (K48)-linked ubiquitin-mediated proteasomal degradation of tripartite motif-containing 47 (TRIM47) and subsequent inactivation of the c-Jun N-terminal kinase (JNK)/p38 mitogen-activated protein kinase (MAPK) cascade are required for the protective function of LMO7 in NASH. CONCLUSIONS These findings provide proof-of-concept evidence supporting LMO7 as a robust suppressor of NASH in the context of hyperlipidemia, indicating that targeting the LMO7-TRIM47 axis is a promising therapeutic strategy for NASH.
Collapse
Affiliation(s)
- Tiangen Wu
- Department of Hepatobiliary & Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, PR China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, Hubei, PR China
| | - Xi Chen
- Department of Hepatobiliary & Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, PR China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, Hubei, PR China
| | - Kequan Xu
- Department of Hepatobiliary & Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, PR China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, Hubei, PR China
| | - Caixia Dai
- Department of Hepatobiliary & Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, PR China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, Hubei, PR China
| | - Xiaomian Li
- Department of Hepatobiliary & Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, PR China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, Hubei, PR China
| | - Yang-Wen-Qing Zhang
- Department of Hepatobiliary & Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, PR China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, Hubei, PR China
| | - Jinghua Li
- Department of Hepatobiliary & Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, PR China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, Hubei, PR China
| | - Meng Gao
- Department of Hepatobiliary & Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, PR China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, Hubei, PR China
| | - Yingyi Liu
- Department of Hepatobiliary & Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, PR China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, Hubei, PR China
| | - Fusheng Liu
- Department of Hepatobiliary & Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, PR China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, Hubei, PR China
| | - Xutao Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, PR China
| | - Bicheng Wang
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, PR China
| | - Peng Xia
- Department of Hepatobiliary & Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, PR China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, Hubei, PR China
| | - Zhen Li
- Department of Hepatobiliary & Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, PR China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, Hubei, PR China
| | - Weijie Ma
- Department of Hepatobiliary & Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, PR China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, Hubei, PR China
| | - Yufeng Yuan
- Department of Hepatobiliary & Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, PR China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, Hubei, PR China
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430071, PR China
| |
Collapse
|
26
|
Roberts MA, Deol KK, Mathiowetz AJ, Lange M, Leto DE, Stevenson J, Hashemi SH, Morgens DW, Easter E, Heydari K, Nalls MA, Bassik MC, Kampmann M, Kopito RR, Faghri F, Olzmann JA. Parallel CRISPR-Cas9 screens identify mechanisms of PLIN2 and lipid droplet regulation. Dev Cell 2023; 58:1782-1800.e10. [PMID: 37494933 PMCID: PMC10530302 DOI: 10.1016/j.devcel.2023.07.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 06/01/2023] [Accepted: 07/03/2023] [Indexed: 07/28/2023]
Abstract
Despite the key roles of perilipin-2 (PLIN2) in governing lipid droplet (LD) metabolism, the mechanisms that regulate PLIN2 levels remain incompletely understood. Here, we leverage a set of genome-edited human PLIN2 reporter cell lines in a series of CRISPR-Cas9 loss-of-function screens, identifying genetic modifiers that influence PLIN2 expression and post-translational stability under different metabolic conditions and in different cell types. These regulators include canonical genes that control lipid metabolism as well as genes involved in ubiquitination, transcription, and mitochondrial function. We further demonstrate a role for the E3 ligase MARCH6 in regulating triacylglycerol biosynthesis, thereby influencing LD abundance and PLIN2 stability. Finally, our CRISPR screens and several published screens provide the foundation for CRISPRlipid (http://crisprlipid.org), an online data commons for lipid-related functional genomics data. Our study identifies mechanisms of PLIN2 and LD regulation and provides an extensive resource for the exploration of LD biology and lipid metabolism.
Collapse
Affiliation(s)
- Melissa A Roberts
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Kirandeep K Deol
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Alyssa J Mathiowetz
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Mike Lange
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Dara E Leto
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Julian Stevenson
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Sayed Hadi Hashemi
- Department of Computer Science, University of Illinois at Urbana-Champaign, Urbana, IL 61820, USA
| | - David W Morgens
- Department of Plant and Microbial Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Emilee Easter
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Kartoosh Heydari
- Cancer Research Laboratory FACS Core Facility, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Mike A Nalls
- Data Tecnica International, LLC, Washington, DC, USA; Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD 20892, USA; Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Michael C Bassik
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Martin Kampmann
- Institute for Neurodegenerative Diseases, Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Ron R Kopito
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Faraz Faghri
- Data Tecnica International, LLC, Washington, DC, USA; Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD 20892, USA; Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - James A Olzmann
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA.
| |
Collapse
|
27
|
Zhang M, Bai X, Du Q, Xu J, Wang D, Chen L, Dong K, Chen Z, Yang J. The Different Mechanisms of Lipid Accumulation in Hepatocytes Induced by Oleic Acid/Palmitic Acid and High-Fat Diet. Molecules 2023; 28:6714. [PMID: 37764494 PMCID: PMC10536454 DOI: 10.3390/molecules28186714] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 09/15/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the primary chronic liver disease worldwide, mainly manifested by hepatic steatosis. Hepatic lipids may be derived from dietary intake, plasma free fatty acid (FFA) uptake, or hepatic de novo lipogenesis (DNL). Currently, cellular and animal models of hepatocellular steatosis are widely used to study the pathogenesis of NAFLD and to investigate therapeutic agents. However, whether there are differences between the in vivo and in vitro models of the mechanisms that cause lipid accumulation has not been reported. We used OA/PA-induced NCTC 1469 cells and high-fat-diet-fed C57BL/6J mice to simulate a hepatocyte steatosis model of NAFLD and to detect indicators related to FFA uptake and DNL. In addition, when serological indicators were analysed in the mouse model, it was found that serum FASN levels decreased. The results revealed that, in the cellular model, indicators related to DNL were decreased, FASN enzyme activity was unchanged, and indicators related to FFA uptake were increased, including the high expression of CD36; while, in the animal model, indicators related to both FFA uptake and de novo synthesis were increased, including the high expression of CD36 and the increased protein levels of FASN with enhanced enzyme activity. In addition, after an analysis of the serological indicators in the mouse model, it was found that the serum levels of FASN were reduced. In conclusion, the OA/PA-induced cellular model can be used to study the mechanism of FFA uptake, whereas the high-fat-diet-induced mouse model can be used to study the mechanism of FFA uptake and DNL. Combined treatment with CD36 and FASN may be more effective against NAFLD. FASN in the serum can be used as one of the indicators for the clinical diagnosis of NAFLD.
Collapse
Affiliation(s)
- Miao Zhang
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing 101400, China; (M.Z.); (X.B.); (Q.D.); (J.X.); (D.W.); (L.C.); (K.D.)
| | - Xue Bai
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing 101400, China; (M.Z.); (X.B.); (Q.D.); (J.X.); (D.W.); (L.C.); (K.D.)
| | - Qian Du
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing 101400, China; (M.Z.); (X.B.); (Q.D.); (J.X.); (D.W.); (L.C.); (K.D.)
| | - Jiaojiao Xu
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing 101400, China; (M.Z.); (X.B.); (Q.D.); (J.X.); (D.W.); (L.C.); (K.D.)
| | - Danqing Wang
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing 101400, China; (M.Z.); (X.B.); (Q.D.); (J.X.); (D.W.); (L.C.); (K.D.)
| | - Lei Chen
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing 101400, China; (M.Z.); (X.B.); (Q.D.); (J.X.); (D.W.); (L.C.); (K.D.)
| | - Keting Dong
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing 101400, China; (M.Z.); (X.B.); (Q.D.); (J.X.); (D.W.); (L.C.); (K.D.)
| | - Ziyue Chen
- School of Nursing, Capital Medical University, Beijing 100069, China;
| | - Jianhong Yang
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing 101400, China; (M.Z.); (X.B.); (Q.D.); (J.X.); (D.W.); (L.C.); (K.D.)
| |
Collapse
|
28
|
Li Q, Wang W, Duan F, Wang Y, Chen S, Shi K, Xia Y, Li X, Gao Y, Liu G. DNMT3B Alleviates Liver Steatosis Induced by Chronic Low-grade LPS via Inhibiting CIDEA Expression. Cell Mol Gastroenterol Hepatol 2023; 17:59-77. [PMID: 37703946 PMCID: PMC10665944 DOI: 10.1016/j.jcmgh.2023.09.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 09/02/2023] [Accepted: 09/05/2023] [Indexed: 09/15/2023]
Abstract
BACKGROUND & AIMS Nonalcoholic fatty liver disease is the most prevalent chronic liver disease and threats to human health. Gut dysbiosis caused by lipopolysaccharide (LPS) leakage has been strongly related to nonalcoholic fatty liver disease progression, although the underlying mechanisms remain unclear. METHODS Previous studies have shown that low-grade LPS administration to mice on a standard, low-fat chow diet is sufficient to induce symptoms of fatty liver. This study confirmed these findings and supported LPS as a lipid metabolism regulator in the liver. RESULTS Mechanically, LPS induced dysregulated lipid metabolism by inhibiting the expression of DNA methyltransferases 3B (DNMT3B). Genetic overexpression of DNMT3B alleviated LPS-induced lipid accumulation, whereas its knockdown increased steatosis in mice and human hepatocytes. LPS-induced lower expression of DNMT3B led to hypomethylation in promoter region of CIDEA, resulting in increased binding of SREBP-1c to its promoter and activated CIDEA expression. Hepatic interference of CIDEA reversed the effect of LPS on lipogenesis. These effects were independent of a high-fat diet or high fatty acid action. CONCLUSIONS Overall, these findings sustain the conclusion that LPS is a lipogenic factor and could be involved in hepatic steatosis progression.
Collapse
Affiliation(s)
- Qiang Li
- Department of Cell Biology, School of Life Science, Bengbu Medical College, Anhui, China; Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Anhui, China.
| | - Wenjing Wang
- Department of Cell Biology, School of Life Science, Bengbu Medical College, Anhui, China
| | - Feifan Duan
- Department of Cell Biology, School of Life Science, Bengbu Medical College, Anhui, China
| | - Yaju Wang
- Department of Cell Biology, School of Life Science, Bengbu Medical College, Anhui, China
| | - Shuya Chen
- Department of Cell Biology, School of Life Science, Bengbu Medical College, Anhui, China
| | - Kangyun Shi
- Department of Cell Biology, School of Life Science, Bengbu Medical College, Anhui, China
| | - Yinyin Xia
- Department of Cell Biology, School of Life Science, Bengbu Medical College, Anhui, China
| | - Xinyu Li
- Department of Cell Biology, School of Life Science, Bengbu Medical College, Anhui, China
| | - Yu Gao
- Department of Cell Biology, School of Life Science, Bengbu Medical College, Anhui, China; Bengbu Medical College Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu Medical College, Bengbu, China
| | - Guoquan Liu
- Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Anhui, China; Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical College, Anhui, China.
| |
Collapse
|
29
|
Bombarda-Rocha V, Silva D, Badr-Eddine A, Nogueira P, Gonçalves J, Fresco P. Challenges in Pharmacological Intervention in Perilipins (PLINs) to Modulate Lipid Droplet Dynamics in Obesity and Cancer. Cancers (Basel) 2023; 15:4013. [PMID: 37568828 PMCID: PMC10417315 DOI: 10.3390/cancers15154013] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/01/2023] [Accepted: 08/04/2023] [Indexed: 08/13/2023] Open
Abstract
Perilipins (PLINs) are the most abundant proteins in lipid droplets (LD). These LD-associated proteins are responsible for upgrading LD from inert lipid storage structures to fully functional organelles, fundamentally integrated in the lipid metabolism. There are five distinct perilipins (PLIN1-5), each with specific expression patterns and metabolic activation, but all capable of regulating the activity of lipases on LD. This plurality creates a complex orchestrated mechanism that is directly related to the healthy balance between lipogenesis and lipolysis. Given the essential role of PLINs in the modulation of the lipid metabolism, these proteins can become interesting targets for the treatment of lipid-associated diseases. Since reprogrammed lipid metabolism is a recognized cancer hallmark, and obesity is a known risk factor for cancer and other comorbidities, the modulation of PLINs could either improve existing treatments or create new opportunities for the treatment of these diseases. Even though PLINs have not been, so far, directly considered for pharmacological interventions, there are many established drugs that can modulate PLINs activity. Therefore, the aim of this study is to assess the involvement of PLINs in diseases related to lipid metabolism dysregulation and whether PLINs can be viewed as potential therapeutic targets for cancer and obesity.
Collapse
Affiliation(s)
- Victória Bombarda-Rocha
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (V.B.-R.); (D.S.); (A.B.-E.); (P.N.); (P.F.)
- UCIBIO–Applied Molecular Biosciences Unit, Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Dany Silva
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (V.B.-R.); (D.S.); (A.B.-E.); (P.N.); (P.F.)
- UCIBIO–Applied Molecular Biosciences Unit, Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Allal Badr-Eddine
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (V.B.-R.); (D.S.); (A.B.-E.); (P.N.); (P.F.)
| | - Patrícia Nogueira
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (V.B.-R.); (D.S.); (A.B.-E.); (P.N.); (P.F.)
- UCIBIO–Applied Molecular Biosciences Unit, Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Jorge Gonçalves
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (V.B.-R.); (D.S.); (A.B.-E.); (P.N.); (P.F.)
- UCIBIO–Applied Molecular Biosciences Unit, Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Paula Fresco
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (V.B.-R.); (D.S.); (A.B.-E.); (P.N.); (P.F.)
- UCIBIO–Applied Molecular Biosciences Unit, Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
30
|
Wu X, Xu M, Geng M, Chen S, Little PJ, Xu S, Weng J. Targeting protein modifications in metabolic diseases: molecular mechanisms and targeted therapies. Signal Transduct Target Ther 2023; 8:220. [PMID: 37244925 PMCID: PMC10224996 DOI: 10.1038/s41392-023-01439-y] [Citation(s) in RCA: 91] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 03/01/2023] [Accepted: 04/06/2023] [Indexed: 05/29/2023] Open
Abstract
The ever-increasing prevalence of noncommunicable diseases (NCDs) represents a major public health burden worldwide. The most common form of NCD is metabolic diseases, which affect people of all ages and usually manifest their pathobiology through life-threatening cardiovascular complications. A comprehensive understanding of the pathobiology of metabolic diseases will generate novel targets for improved therapies across the common metabolic spectrum. Protein posttranslational modification (PTM) is an important term that refers to biochemical modification of specific amino acid residues in target proteins, which immensely increases the functional diversity of the proteome. The range of PTMs includes phosphorylation, acetylation, methylation, ubiquitination, SUMOylation, neddylation, glycosylation, palmitoylation, myristoylation, prenylation, cholesterylation, glutathionylation, S-nitrosylation, sulfhydration, citrullination, ADP ribosylation, and several novel PTMs. Here, we offer a comprehensive review of PTMs and their roles in common metabolic diseases and pathological consequences, including diabetes, obesity, fatty liver diseases, hyperlipidemia, and atherosclerosis. Building upon this framework, we afford a through description of proteins and pathways involved in metabolic diseases by focusing on PTM-based protein modifications, showcase the pharmaceutical intervention of PTMs in preclinical studies and clinical trials, and offer future perspectives. Fundamental research defining the mechanisms whereby PTMs of proteins regulate metabolic diseases will open new avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Xiumei Wu
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, Anhui, 230001, China
- Department of Endocrinology and Metabolism, Guangdong Provincial Key Laboratory of Diabetology, The Third Affiliated Hospital of Sun Yat-sen University, 510000, Guangzhou, China
| | - Mengyun Xu
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Mengya Geng
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Shuo Chen
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Peter J Little
- School of Pharmacy, University of Queensland, Pharmacy Australia Centre of Excellence, Woolloongabba, QLD, 4102, Australia
- Sunshine Coast Health Institute and School of Health and Behavioural Sciences, University of the Sunshine Coast, Birtinya, QLD, 4575, Australia
| | - Suowen Xu
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Jianping Weng
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, Anhui, 230001, China.
- Department of Endocrinology and Metabolism, Guangdong Provincial Key Laboratory of Diabetology, The Third Affiliated Hospital of Sun Yat-sen University, 510000, Guangzhou, China.
- Bengbu Medical College, Bengbu, 233000, China.
| |
Collapse
|
31
|
Xu J, Liu Z, Zhang J, Chen S, Wang W, Zhao X, Zhen M, Huang X. N-end Rule-Mediated Proteasomal Degradation of ATGL Promotes Lipid Storage. Diabetes 2023; 72:210-222. [PMID: 36346641 PMCID: PMC9871197 DOI: 10.2337/db22-0362] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022]
Abstract
Cellular lipid storage is regulated by the balance of lipogenesis and lipolysis. The rate-limiting triglyceride hydrolase ATGL (desnutrin/PNPLA2) is critical for lipolysis. The control of ATGL transcription, localization, and activation has been intensively studied, while regulation of the protein stability of ATGL is much less explored. In this study, we showed that the protein stability of ATGL is regulated by the N-end rule in cultured cells and in mice. The N-end rule E3 ligases UBR1 and UBR2 reduce the level of ATGL and affect lipid storage. The N-end rule-resistant ATGL(F2A) mutant, in which the N-terminal phenylalanine (F) of ATGL is substituted by alanine (A), has increased protein stability and enhanced lipolysis activity. ATGLF2A/F2A knock-in mice are protected against high-fat diet (HFD)-induced obesity, hepatic steatosis, and insulin resistance. Hepatic knockdown of Ubr1 attenuates HFD-induced hepatic steatosis by enhancing the ATGL level. Finally, the protein levels of UBR1 and ATGL are negatively correlated in the adipose tissue of obese mice. Our study reveals N-end rule-mediated proteasomal regulation of ATGL, a finding that may potentially be beneficial for treatment of obesity.
Collapse
Affiliation(s)
- Jiesi Xu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- Corresponding authors: Jiesi Xu, , and Xun Huang,
| | - Zhenglong Liu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jianxin Zhang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Siyu Chen
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Wei Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Xuefan Zhao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Mei Zhen
- Lunenfeld–Tanebaum Research Institute, Departments of Molecular Genetics and Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Xun Huang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- Corresponding authors: Jiesi Xu, , and Xun Huang,
| |
Collapse
|
32
|
Zhu M, Wang X, Wang K, Zhao Z, Dang Y, Ji G, Li F, Zhou W. Lingguizhugan decoction improves non-alcoholic steatohepatitis partially by modulating gut microbiota and correlated metabolites. Front Cell Infect Microbiol 2023; 13:1066053. [PMID: 36779187 PMCID: PMC9908757 DOI: 10.3389/fcimb.2023.1066053] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 01/12/2023] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Lingguizhugan decoction is a traditional Chinese medicine prescription that has been used to improve non-alcoholic fatty liver disease and its progressive form, non-alcoholic steatohepatitis (NASH). However, the anti-NASH effects and underlying mechanisms of Lingguizhugan decoction remain unclear. METHODS Male Sprague-Dawley rats were fed a methionine- and choline-deficient (MCD) diet to induce NASH, and then given Lingguizhugan decoction orally for four weeks. NASH indexes were evaluated by histopathological analysis and biochemical parameters including serum alanine aminotransferase (ALT), aspartate aminotransferase (AST), liver triglycerides (TG), etc. Fecal samples of rats were subjected to profile the changes of gut microbiota and metabolites using 16S rRNA sequencing and ultra-performance liquid chromatography coupled to tandem mass spectrometry (UPLC-MS). Bioinformatics was used to identify Lingguizhugan decoction reversed candidates, and Spearman's correlation analysis was performed to uncover the relationship among gut microbiota, fecal metabolites, and NASH indexes. RESULTS Four-week Lingguizhugan decoction treatment ameliorated MCD diet-induced NASH features, as evidenced by improved hepatic steatosis and inflammation, as well as decreased serum AST and ALT levels. Besides, Lingguizhugan decoction partially restored the changes in gut microbial community composition in NASH rats. Meanwhile, the relative abundance of 26 genera was significantly changed in NASH rats, and 11 genera (such as odoribacter, Ruminococcus_1, Ruminococcaceae_UCG-004, etc.) were identified as significantly reversed by Lingguizhugan decoction. Additionally, a total of 99 metabolites were significantly altered in NASH rats, and 57 metabolites (such as TDCA, Glutamic acid, Isocaproic acid, etc.) enriched in different pathways were reversed by Lingguizhugan decoction. Furthermore, Spearman's correlation analyses revealed that most of the 57 metabolites were significantly correlated with 11 genera and NASH indexes. CONCLUSION Lingguizhugan decoction may exert protective effects on NASH partially by modulating gut microbiota and correlated metabolites.
Collapse
Affiliation(s)
- Mingzhe Zhu
- Institute of Digestive Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- School of Public Health, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xue Wang
- Institute of Digestive Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Kai Wang
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhiqiang Zhao
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yanqi Dang
- Institute of Digestive Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guang Ji
- Institute of Digestive Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fenghua Li
- Institute of Digestive Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wenjun Zhou
- Institute of Digestive Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
33
|
Li X, Shan K, Li C, Zhou G. Intermittent Protein Diets Alter Hepatic Lipid Accumulation by Changing Tryptophan Metabolism in a Fast-Response Manner. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:3261-3272. [PMID: 36634216 DOI: 10.1021/acs.jafc.2c06576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
In modern life, the fluctuation of dietary protein levels is common, in particular, for low-income populations. However, its effect on human health is little known. Alternating changes of low and high casein or pork protein were used to simulate the fluctuation of dietary protein content in mice. Hepatic lipid accumulation showed a fast response to alternating changes of low- and high-protein diets. Correspondingly, some gut microbiota and tryptophan metabolite composition also showed a fast response to dietary protein changes. The fast response of 3-hydroxykynurenine (3-HK) was proven to inhibit hepatic lipid accumulation in vitro. Therefore, intermittent protein diets modulated hepatic lipid accumulation through 3-HK. These findings highlighted the sensitivity of hepatic lipid accumulation to dietary protein levels.
Collapse
Affiliation(s)
- Xiaohui Li
- Key Laboratory of Meat Processing and Quality Control, Ministry of Education; Key Laboratory of Meat Processing, Ministry of Agriculture and Rural Affairs; Jiangsu Synergistic Innovation Center of Meat Processing and Quality Control, College of Food Science and Technology, Nanjing Agricultural University, 210095Nanjing, P. R. China
| | - Kai Shan
- Key Laboratory of Meat Processing and Quality Control, Ministry of Education; Key Laboratory of Meat Processing, Ministry of Agriculture and Rural Affairs; Jiangsu Synergistic Innovation Center of Meat Processing and Quality Control, College of Food Science and Technology, Nanjing Agricultural University, 210095Nanjing, P. R. China
| | - Chunbao Li
- Key Laboratory of Meat Processing and Quality Control, Ministry of Education; Key Laboratory of Meat Processing, Ministry of Agriculture and Rural Affairs; Jiangsu Synergistic Innovation Center of Meat Processing and Quality Control, College of Food Science and Technology, Nanjing Agricultural University, 210095Nanjing, P. R. China
| | - Guanghong Zhou
- Key Laboratory of Meat Processing and Quality Control, Ministry of Education; Key Laboratory of Meat Processing, Ministry of Agriculture and Rural Affairs; Jiangsu Synergistic Innovation Center of Meat Processing and Quality Control, College of Food Science and Technology, Nanjing Agricultural University, 210095Nanjing, P. R. China
| |
Collapse
|
34
|
Shao C, Xu L, Lei P, Wang W, Feng S, Ye J, Zhong B. Metabolomics to identify fingerprints of carotid atherosclerosis in nonobese metabolic dysfunction-associated fatty liver disease. J Transl Med 2023; 21:12. [PMID: 36624524 PMCID: PMC9830861 DOI: 10.1186/s12967-022-03760-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/06/2022] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND/AIMS Nonobese metabolic dysfunction-associated fatty liver disease (MAFLD) is paradoxically associated with improved metabolic and pathological features at diagnosis but similar cardiovascular diseases (CVD) prognosis to obese MAFLD. We aimed to utilize the metabolomics to identify the potential metabolite profiles accounting for this phenomenon. METHODS This prospective multicenter cross-sectional study was conducted in China enrolling derivation and validation cohorts. Liquid chromatography coupled with mass spectrometry and gas chromatography-mass spectrometry were applied to perform a metabolomics measurement. RESULTS The study involved 120 MAFLD patients and 60 non-MAFLD controls in the derivation cohort. Controls were divided into two groups according to the presence of carotid atherosclerosis (CAS). The MAFLD group was further divided into nonobese MAFLD with/without CAS groups and obese MAFLD with/without CAS groups. Fifty-six metabolites were statistically significant for discriminating the six groups. Among the top 10 metabolites related to CAS in nonobese MAFLD, only phosphatidylethanolamine (PE 20:2/16:0), phosphatidylglycerol (PG 18:0/20:4) and de novo lipogenesis (16:0/18:2n-6) achieved significant areas under the ROC curve (AUCs, 0.67, p = 0.03; 0.79, p = 0.02; 0.63, p = 0.03, respectively). The combination of these three metabolites and liver stiffness achieved a significantly higher AUC (0.92, p < 0.01). In obese MAFLD patients, cystine was found to be significant with an AUC of 0.69 (p = 0.015), followed by sphingomyelin (SM 16:1/18:1) (0.71, p = 0.004) and de novo lipogenesis (16:0/18:2n-6) (0.73, p = 0.004). The combination of these three metabolites, liver fat content and age attained a significantly higher AUC of 0.91 (p < 0.001). The AUCs of these metabolites remained highly significant in the independent validation cohorts involving 200 MAFLD patients and 90 controls. CONCLUSIONS Diagnostic models combining different metabolites according to BMI categories could raise the accuracy of identifying subclinical CAS. Trial registration The study protocol was approved by the local ethics committee and all the participants have provided written informed consent (Approval number: [2014] No. 112, registered at the Chinese Clinical Trial Registry, ChiCTR-ChiCTR2000034197).
Collapse
Affiliation(s)
- Congxiang Shao
- grid.12981.330000 0001 2360 039XDepartment of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, No. 58 Zhongshan II Road, Yuexiu District, Guangzhou, 510080 China
| | - Lishu Xu
- grid.410643.4Department of Gastroenterology and Hepatology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, and Guangdong Provincial Geriatrics Institute, No. 106 Zhongshan II Road, Yuexiu District, Guangzhou, China
| | - Pingguang Lei
- Department of Gastroenterology, Shenzhen Baoan District Songgang People’s Hospital, No. 2, Shajiang Road, Songgang Street, Bao’an District, Shenzhen, China
| | - Wei Wang
- grid.12981.330000 0001 2360 039XDepartment of Medical Ultrasonics, The First Affiliated Hospital, Sun Yat-Sen University, No. 58 Zhongshan II Road, Yuexiu District, Guangzhou, China
| | - Shiting Feng
- grid.12981.330000 0001 2360 039XDepartment of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, No. 58 Zhongshan II Road, Yuexiu District, Guangzhou, China
| | - Junzhao Ye
- grid.12981.330000 0001 2360 039XDepartment of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, No. 58 Zhongshan II Road, Yuexiu District, Guangzhou, 510080 China
| | - Bihui Zhong
- grid.12981.330000 0001 2360 039XDepartment of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, No. 58 Zhongshan II Road, Yuexiu District, Guangzhou, 510080 China
| |
Collapse
|
35
|
Sherpa D, Chrustowicz J, Schulman BA. How the ends signal the end: Regulation by E3 ubiquitin ligases recognizing protein termini. Mol Cell 2022; 82:1424-1438. [PMID: 35247307 PMCID: PMC9098119 DOI: 10.1016/j.molcel.2022.02.004] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/18/2022] [Accepted: 02/01/2022] [Indexed: 12/31/2022]
Abstract
Specificity of eukaryotic protein degradation is determined by E3 ubiquitin ligases and their selective binding to protein motifs, termed "degrons," in substrates for ubiquitin-mediated proteolysis. From the discovery of the first substrate degron and the corresponding E3 to a flurry of recent studies enabled by modern systems and structural methods, it is clear that many regulatory pathways depend on E3s recognizing protein termini. Here, we review the structural basis for recognition of protein termini by E3s and how this recognition underlies biological regulation. Diverse E3s evolved to harness a substrate's N and/or C terminus (and often adjacent residues as well) in a sequence-specific manner. Regulation is achieved through selective activation of E3s and also through generation of degrons at ribosomes or by posttranslational means. Collectively, many E3 interactions with protein N and C termini enable intricate control of protein quality and responses to cellular signals.
Collapse
Affiliation(s)
- Dawafuti Sherpa
- Department of Molecular Machines and Signaling, Max Planck Institute of Biochemistry, Martinsried, Bavaria, Germany
| | - Jakub Chrustowicz
- Department of Molecular Machines and Signaling, Max Planck Institute of Biochemistry, Martinsried, Bavaria, Germany
| | - Brenda A Schulman
- Department of Molecular Machines and Signaling, Max Planck Institute of Biochemistry, Martinsried, Bavaria, Germany.
| |
Collapse
|