1
|
Zhao X, Wang S, Liu Q, Wei W, Sun X, Song H, Xu J, Zhang S, Wang H, Fu J. Single-cell landscape of the intrahepatic ecosystem in alcohol-related liver disease. Clin Transl Med 2025; 15:e70198. [PMID: 39834100 PMCID: PMC11746962 DOI: 10.1002/ctm2.70198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 01/05/2025] [Accepted: 01/10/2025] [Indexed: 01/22/2025] Open
Abstract
Alcohol-related liver disease (ALD) is a common chronic liver disease caused by long-term excessive alcohol consumption and responsible for more than half of all liver-related deaths worldwide. The molecular mechanisms associated with ALD were not fully understood. In this study, we performed single-cell RNA sequencing on liver tissues obtained from ALD patients and healthy liver donors. We identified an ALB+KRT7+ epithelial population that expressed both hepatocyte and biliary markers significantly expanded in ALD livers. The ALB+KRT7+ epithelial cells were demonstrated to have stem cell properties and malignant transformation potentials. Moreover, ALB+KRT7+ epithelium-derived ALD organoids promote the tumour growth by activating Wnt/β-catenin signalling of liver cancer cells. Most importantly, blocking the Wnt protein secretion or knockdown the Wnt receptor suppressed the tumour promoting effect of ALD organoids. Our study provides important insights that Wnt signalling can be targeted in patients with advanced alcohol-related cirrhosis to prevent malignant transformation. In addition, our results also uncovered the important alterations of nonparenchymal cells, especially macrophages and T/NK populations that responsible for active inflammation responses in alcohol-related hepatitis and immunosuppressive microenvironment in advanced cirrhosis livers, which likely facilitated the malignant progression of ALD. KEY POINTS: This study provides single-cell landscape of human liver samples across different ALD stages. The ALB+ KRT7+ epithelium were enriched in ALD patients, and the function of this epithelial population varied significantly across ALD stages. ALB+KRT7+ epithelium from advanced alcohol-related cirrhosis had malignant transformation potential and tumour promotion activity. The comprehensive changes of parenchymal and nonparenchymal cells in the ALD livers lay a hidden danger for the further malignant progression.
Collapse
Affiliation(s)
- Xiaofang Zhao
- Translational Medicine CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- International Cooperation Laboratory on Signal Transduction, National Center for Liver Cancer, Ministry of Education Key Laboratory on Signaling Regulation and Targeting Therapy of Liver Cancer, Shanghai Key Laboratory of Hepatobiliary Tumor BiologyEastern Hepatobiliary Surgery Hospital, Second Military Medical University/NAVAL Medical UniversityShanghaiChina
| | - Senyan Wang
- Translational Medicine CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Qi Liu
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Wenjuan Wei
- Translational Medicine CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Xiaoyan Sun
- Translational Medicine CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Hao Song
- Translational Medicine CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Jing Xu
- Translational Medicine CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Shuijun Zhang
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Hongyang Wang
- International Cooperation Laboratory on Signal Transduction, National Center for Liver Cancer, Ministry of Education Key Laboratory on Signaling Regulation and Targeting Therapy of Liver Cancer, Shanghai Key Laboratory of Hepatobiliary Tumor BiologyEastern Hepatobiliary Surgery Hospital, Second Military Medical University/NAVAL Medical UniversityShanghaiChina
| | - Jing Fu
- International Cooperation Laboratory on Signal Transduction, National Center for Liver Cancer, Ministry of Education Key Laboratory on Signaling Regulation and Targeting Therapy of Liver Cancer, Shanghai Key Laboratory of Hepatobiliary Tumor BiologyEastern Hepatobiliary Surgery Hospital, Second Military Medical University/NAVAL Medical UniversityShanghaiChina
| |
Collapse
|
2
|
Reid MV, Fredickson G, Mashek DG. Mechanisms coupling lipid droplets to MASLD pathophysiology. Hepatology 2024:01515467-990000000-01067. [PMID: 39475114 DOI: 10.1097/hep.0000000000001141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 10/17/2024] [Indexed: 01/03/2025]
Abstract
Hepatic steatosis, the buildup of neutral lipids in lipid droplets (LDs), is commonly referred to as metabolic dysfunction-associated steatotic liver disease when alcohol or viral infections are not involved. Metabolic dysfunction-associated steatotic liver disease encompasses simple steatosis and the more severe metabolic dysfunction-associated steatohepatitis, characterized by inflammation, hepatocyte injury, and fibrosis. Previously viewed as inert markers of disease, LDs are now understood to play active roles in disease etiology and have significant nonpathological and pathological functions in cell signaling and function. These dynamic properties of LDs are tightly regulated by hundreds of proteins that coat the LD surface, controlling lipid metabolism, trafficking, and signaling. The following review highlights various facets of LD biology with the primary goal of discussing key mechanisms through which LDs promote the development of advanced liver diseases, including metabolic dysfunction-associated steatohepatitis.
Collapse
Affiliation(s)
- Mari V Reid
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Gavin Fredickson
- Department of Integrated Biology and Physiology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Douglas G Mashek
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, University of Minnesota, Minneapolis, Minnesota, USA
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
3
|
Huang JH, Wei Y, Fang Z, Yu C, Zhang R, Feng ZB, Zeng LP. Clinical pathological significance and biological function of PLIN1 in hepatocellular carcinoma: bioinformatics analysis and in vitro experiments. BMC Cancer 2024; 24:1073. [PMID: 39215210 PMCID: PMC11363539 DOI: 10.1186/s12885-024-12842-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND & AIMS Perilipin 1 (PLIN1) is an essential lipid droplet surface protein that participates in cell life activities by regulating energy balance and lipid metabolism. PLIN1 has been shown to be closely related to the development of numerous tumor types. The purpose of this work was to elucidate the clinicopathologic significance of PLIN1 in hepatocellular carcinoma (HCC), as well as its impact on the biological functions of HCC cells, and to investigate the underlying mechanisms involved. METHODS Public high-throughput RNA microarray and RNA sequencing data were collected to examine PLIN1 levels and clinical significance in patients with HCC. Immunohistochemistry (IHC) and real-time quantitative reverse transcription polymerase chain reaction (RT‒qPCR) were conducted to assess the expression levels and the clinicopathological relevance of PLIN1 in HCC. Then, SK and Huh7 cells were transfected with a lentivirus overexpressing PLIN1. CCK8 assay, wound healing assay, transwell assay, and flow cytometric analysis were conducted to explore the effects of PLIN1 overexpression on HCC cell proliferation, migration, invasion, and cell cycle distribution. Ultimately, Gene Ontology (GO) functional annotation and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis were performed to investigate the underlying mechanisms of PLIN1 in HCC progression based on HCC differentially expressed genes and PLIN1 co-expressed genes. RESULTS PLIN1 was markedly downregulated in HCC tissues, which correlated with a noticeably worse prognosis for HCC patients. Additionally, PLIN1 overexpression inhibited the proliferation, migration, and invasion of SK and Huh7 cells in vitro, as well as arresting the HCC cell cycle at the G0/G1 phase. More significantly, energy conversion-related biological processes, lipid metabolism, and cell cycle signalling pathways were the three most enriched molecular mechanisms. CONCLUSION The present study revealed that PLIN1 downregulation is associated with poor prognosis in HCC patients and accelerated HCC progression by promoting cellular proliferation, migration, and metastasis, as well as the mechanisms underlying the regulation of lipid metabolism-related pathways in HCC.
Collapse
Affiliation(s)
- Jiang-Hua Huang
- Department of Pathology, Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou Worker's Hospital, Liuzhou, Guangxi Zhuang Autonomous Region, 545000, People's Republic of China
| | - Yan Wei
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, People's Republic of China
| | - Zhen Fang
- Department of Pathology, Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou Worker's Hospital, Liuzhou, Guangxi Zhuang Autonomous Region, 545000, People's Republic of China
| | - Cong Yu
- Department of Pathology, Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region , 530000, China
| | - Rui Zhang
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, People's Republic of China
| | - Zhen-Bo Feng
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, People's Republic of China.
| | - Li-Ping Zeng
- Department of Pathology, Hunan University of Medicine, 492 Jinxinan RD, Huaihua, Hunan, 418000, People's Republic of China.
| |
Collapse
|
4
|
Tiniakos DG, Anstee QM, Brunt EM, Burt AD. Fatty Liver Disease. MACSWEEN'S PATHOLOGY OF THE LIVER 2024:330-401. [DOI: 10.1016/b978-0-7020-8228-3.00005-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
5
|
Mastoridou EM, Goussia AC, Kanavaros P, Charchanti AV. Involvement of Lipophagy and Chaperone-Mediated Autophagy in the Pathogenesis of Non-Alcoholic Fatty Liver Disease by Regulation of Lipid Droplets. Int J Mol Sci 2023; 24:15891. [PMID: 37958873 PMCID: PMC10649352 DOI: 10.3390/ijms242115891] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 10/30/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is defined as the accumulation of lipids in the form of lipid droplets in more than 5% of hepatocytes. It is regarded as a range of diverse pathologies, including simple steatosis and steatohepatitis. The structural characteristics of lipid droplets, along with their protein composition, mainly including perilipins, have been implicated in the etiology of the disease. These proteins have garnered increasing attention as a pivotal regulator since their levels and distinct expression appear to be associated with the progression from simple steatosis to steatohepatitis. Perilipins are target proteins of chaperone-mediated autophagy, and their degradation is a prerequisite for lipolysis and lipophagy to access the lipid core. Both lipophagy and chaperone-mediated autophagy have significant implications on the development of the disease, as evidenced by their upregulation during the initial phases of simple steatosis and their subsequent downregulation once steatosis is established. On the contrary, during steatohepatitis, the process of chaperone-mediated autophagy is enhanced, although lipophagy remains suppressed. Evidently, the reduced levels of autophagic pathways observed in simple steatosis serve as a defensive mechanism against lipotoxicity. Conversely, in steatohepatitis, chaperone-mediated autophagy fails to compensate for the continuous generation of small lipid droplets and thus cannot protect hepatocytes from lipotoxicity.
Collapse
Affiliation(s)
- Eleftheria M. Mastoridou
- Department of Anatomy-Histology-Embryology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (E.M.M.); (P.K.)
| | - Anna C. Goussia
- Department of Pathology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece;
| | - Panagiotis Kanavaros
- Department of Anatomy-Histology-Embryology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (E.M.M.); (P.K.)
| | - Antonia V. Charchanti
- Department of Anatomy-Histology-Embryology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (E.M.M.); (P.K.)
| |
Collapse
|
6
|
Wang S, Zhou Y, Yu R, Ling J, Li B, Yang C, Cheng Z, Qian R, Lin Z, Yu C, Zheng J, Zheng X, Jia Q, Wu W, Wu Q, Chen M, Yuan S, Dong W, Shi Y, Jansen R, Yang C, Hao Y, Yao M, Qin W, Jin H. Loss of hepatic FTCD promotes lipid accumulation and hepatocarcinogenesis by upregulating PPARγ and SREBP2. JHEP Rep 2023; 5:100843. [PMID: 37675273 PMCID: PMC10477690 DOI: 10.1016/j.jhepr.2023.100843] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 06/02/2023] [Accepted: 06/26/2023] [Indexed: 09/08/2023] Open
Abstract
Background & Aims Exploiting key regulators responsible for hepatocarcinogenesis is of great importance for the prevention and treatment of hepatocellular carcinoma (HCC). However, the key players contributing to hepatocarcinogenesis remain poorly understood. We explored the molecular mechanisms underlying the carcinogenesis and progression of HCC for the development of potential new therapeutic targets. Methods The Cancer Genome Atlas-Liver Hepatocellular Carcinoma (TCGA-LIHC) and Genotype-Tissue Expression (GTEx) databases were used to identify genes with enhanced expression in the liver associated with HCC progression. A murine liver-specific Ftcd knockout (Ftcd-LKO) model was generated to investigate the role of formimidoyltransferase cyclodeaminase (FTCD) in HCC. Multi-omics analysis of transcriptomics, metabolomics, and proteomics data were applied to further analyse the molecular effects of FTCD expression on hepatocarcinogenesis. Functional and biochemical studies were performed to determine the significance of loss of FTCD expression and the therapeutic potential of Akt inhibitors in FTCD-deficient cancer cells. Results FTCD is highly expressed in the liver but significantly downregulated in HCC. Patients with HCC and low levels of FTCD exhibited worse prognosis, and patients with liver cirrhosis and low FTCD levels exhibited a notable higher probability of developing HCC. Hepatocyte-specific knockout of FTCD promoted both chronic diethylnitrosamine-induced and spontaneous hepatocarcinogenesis in mice. Multi-omics analysis showed that loss of FTCD affected fatty acid and cholesterol metabolism in hepatocarcinogenesis. Mechanistically, loss of FTCD upregulated peroxisome proliferator-activated receptor (PPAR)γ and sterol regulatory element-binding protein 2 (SREBP2) by regulating the PTEN/Akt/mTOR signalling axis, leading to lipid accumulation and hepatocarcinogenesis. Conclusions Taken together, we identified a FTCD-regulated lipid metabolic mechanism involving PPARγ and SREBP2 signaling in hepatocarcinogenesis and provide a rationale for therapeutically targeting of HCC driven by downregulation of FTCD. Impact and implications Exploiting key molecules responsible for hepatocarcinogenesis is significant for the prevention and treatment of HCC. Herein, we identified formimidoyltransferase cyclodeaminase (FTCD) as the top enhanced gene, which could serve as a predictive and prognostic marker for patients with HCC. We generated and characterised the first Ftcd liver-specific knockout murine model. We found loss of FTCD expression upregulated peroxisome proliferator-activated receptor (PPAR)γ and sterol regulatory element-binding protein 2 (SREBP2) by regulating the PTEN/Akt/mTOR signalling axis, leading to lipid accumulation and hepatocarcinogenesis, and provided a rationale for therapeutic targeting of HCC driven by downregulation of FTCD.
Collapse
Affiliation(s)
- Siying Wang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yangyang Zhou
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruobing Yu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Ling
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Botai Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Yang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhuoan Cheng
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ruolan Qian
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhang Lin
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chengtao Yu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiaojiao Zheng
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xingling Zheng
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qi Jia
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Wu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiangxin Wu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mengnuo Chen
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shengxian Yuan
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| | - Wei Dong
- Department of Pathology, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, Shanghai, China
| | - Yaoping Shi
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Robin Jansen
- Division of Molecular Carcinogenesis, Oncode Institute, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Chen Yang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Pharmaceutical Co. Ltd., Nanjing, China
| | - Yujun Hao
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ming Yao
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenxin Qin
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haojie Jin
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
7
|
Yue C, Li D, Fan S, Tao F, Yu Y, Lu W, Chen Q, Yuan A, Wu J, Zhao G, Dong H, Hu Y. Long-term and liver-selected ginsenoside C-K nanoparticles retard NAFLD progression by restoring lipid homeostasis. Biomaterials 2023; 301:122291. [PMID: 37619263 DOI: 10.1016/j.biomaterials.2023.122291] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 07/25/2023] [Accepted: 08/19/2023] [Indexed: 08/26/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most prevalent hepatic disease characterized as lipid accumulation, yet without any approved drug. And development of therapeutic molecules is obstructed by low efficiency and organ toxicity. Herein, we develop a long-term, low-toxic and liver-selected nano candidate, nabCK, to alleviate NAFLD. NabCK is simply composed by natural compound ginsenoside compound K (CK) and albumin. As a major metabolite of ginseng, ginsenoside CK has excellently modulating functions for lipid metabolism, but accompanied by an extremely poor bioavailability <1%. Albumin is a key lipid carrier secreted and metabolized by livers. Thereby, it can improve solubility and liver-localization of CK. In adipocytes and hepatocytes, nabCK prevents lipid deposition and eliminates lipid droplets. Transcriptomic analysis reveals that nabCK rectifies various pathways that involved in steatosis development, including lipid absorption, lipid export, fatty acid biosynthesis, lipid storage and inflammation. All these pathways are modulated by mTOR, the pivotal feedback sensor that is hyperactive in NAFLD. NabCK suppresses mTOR activation to restores lipid homeostasis. In high-fat diet (HFD) induced NAFLD mice, nabCK retards development of steatosis and fibrosis, coupling a protective effect on cardiac tissues from lipotoxicity. Together, nabCK is a safe and potent candidate to offer benefits for NAFLD treatment.
Collapse
Affiliation(s)
- Chunyan Yue
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, School of Life Science, Nanjing University, Nanjing , 210093, China; Institute of Drug R&D, Medical School, Nanjing University, Nanjing, 210093, China
| | - Dandan Li
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, School of Life Science, Nanjing University, Nanjing , 210093, China; Institute of Drug R&D, Medical School, Nanjing University, Nanjing, 210093, China
| | - Shuxin Fan
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, School of Life Science, Nanjing University, Nanjing , 210093, China; Institute of Drug R&D, Medical School, Nanjing University, Nanjing, 210093, China
| | - Feng Tao
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, School of Life Science, Nanjing University, Nanjing , 210093, China; Institute of Drug R&D, Medical School, Nanjing University, Nanjing, 210093, China
| | - Yue Yu
- Affiliated Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, China
| | - Wenjing Lu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, School of Life Science, Nanjing University, Nanjing , 210093, China; Institute of Drug R&D, Medical School, Nanjing University, Nanjing, 210093, China
| | - Qian Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, School of Life Science, Nanjing University, Nanjing , 210093, China; Institute of Drug R&D, Medical School, Nanjing University, Nanjing, 210093, China
| | - Ahu Yuan
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, School of Life Science, Nanjing University, Nanjing , 210093, China; Institute of Drug R&D, Medical School, Nanjing University, Nanjing, 210093, China
| | - Jinhui Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, School of Life Science, Nanjing University, Nanjing , 210093, China; Institute of Drug R&D, Medical School, Nanjing University, Nanjing, 210093, China
| | - Guoping Zhao
- Key Laboratory of Synthetic Biology, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200032, China
| | - Hong Dong
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, School of Life Science, Nanjing University, Nanjing , 210093, China; Institute of Drug R&D, Medical School, Nanjing University, Nanjing, 210093, China.
| | - Yiqiao Hu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, School of Life Science, Nanjing University, Nanjing , 210093, China; Institute of Drug R&D, Medical School, Nanjing University, Nanjing, 210093, China.
| |
Collapse
|
8
|
Yuan Z, Lu X, Lei F, Sun H, Jiang J, Xing D, Du L. Novel Effect of p-Coumaric Acid on Hepatic Lipolysis: Inhibition of Hepatic Lipid-Droplets. Molecules 2023; 28:4641. [PMID: 37375195 DOI: 10.3390/molecules28124641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 06/03/2023] [Accepted: 06/05/2023] [Indexed: 06/29/2023] Open
Abstract
p-coumaric acid (p-CA), a common plant phenolic acid with multiple bioactivities, has a lipid-lowering effect. As a dietary polyphenol, its low toxicity, with the advantages of prophylactic and long-term administration, makes it a potential drug for prophylaxis and the treatment of nonalcoholic fatty liver disease (NAFLD). However, the mechanism by which it regulates lipid metabolism is still unclear. In this study, we studied the effect of p-CA on the down-regulation of accumulated lipids in vivo and in vitro. p-CA increased a number of lipase expressions, including hormone-sensitive lipase (HSL), monoacylglycerol lipase (MGL) and hepatic triglyceride lipase (HTGL), as well as the expression of genes related to fatty acid oxidation, including long-chain fatty acyl-CoA synthetase 1 (ACSL1), carnitine palmitoyltransferase-1 (CPT1), by activating peroxisome proliferator-activated receptor α, and γ (PPARα and γ). Furthermore, p-CA promoted adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) phosphorylation and enhanced the expression of the mammalian suppressor of Sec4 (MSS4), a critical protein that can inhibit lipid droplet growth. Thus, p-CA can decrease lipid accumulation and inhibit lipid droplet fusion, which are correlated with the enhancement of liver lipases and genes related to fatty acid oxidation as an activator of PPARs. Therefore, p-CA is capable of regulating lipid metabolism and is a potential therapeutic drug or health care product for hyperlipidemia and fatty liver.
Collapse
Affiliation(s)
- Zhiyi Yuan
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Xi Lu
- School of Life Sciences and School of Medicine, Tsinghua University, Beijing 100084, China
| | - Fan Lei
- School of Life Sciences and School of Medicine, Tsinghua University, Beijing 100084, China
| | - Hong Sun
- Institute of Medicinal Plant and Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100094, China
| | - Jingfei Jiang
- School of Life Sciences and School of Medicine, Tsinghua University, Beijing 100084, China
| | - Dongming Xing
- School of Life Sciences and School of Medicine, Tsinghua University, Beijing 100084, China
| | - Lijun Du
- School of Life Sciences and School of Medicine, Tsinghua University, Beijing 100084, China
| |
Collapse
|
9
|
Deprince A, Hennuyer N, Kooijman S, Pronk ACM, Baugé E, Lienard V, Verrijken A, Dirinck E, Vonghia L, Woitrain E, Kloosterhuis NJ, Marez E, Jacquemain P, Wolters JC, Lalloyer F, Eberlé D, Quemener S, Vallez E, Tailleux A, Kouach M, Goossens J, Raverdy V, Derudas B, Kuivenhoven JA, Croyal M, van de Sluis B, Francque S, Pattou F, Rensen PCN, Staels B, Haas JT. Apolipoprotein F is reduced in humans with steatosis and controls plasma triglyceride-rich lipoprotein metabolism. Hepatology 2023; 77:1287-1302. [PMID: 35735979 PMCID: PMC10026963 DOI: 10.1002/hep.32631] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 05/19/2022] [Accepted: 06/07/2022] [Indexed: 12/08/2022]
Abstract
BACKGROUND NAFLD affects nearly 25% of the global population. Cardiovascular disease (CVD) is the most common cause of death among patients with NAFLD, in line with highly prevalent dyslipidemia in this population. Increased plasma triglyceride (TG)-rich lipoprotein (TRL) concentrations, an important risk factor for CVD, are closely linked with hepatic TG content. Therefore, it is of great interest to identify regulatory mechanisms of hepatic TRL production and remnant uptake in the setting of hepatic steatosis. APPROACH AND RESULTS To identify liver-regulated pathways linking intrahepatic and plasma TG metabolism, we performed transcriptomic analysis of liver biopsies from two independent cohorts of obese patients. Hepatic encoding apolipoprotein F ( APOF ) expression showed the fourth-strongest negatively correlation with hepatic steatosis and the strongest negative correlation with plasma TG levels. The effects of adenoviral-mediated human ApoF (hApoF) overexpression on plasma and hepatic TG were assessed in C57BL6/J mice. Surprisingly, hApoF overexpression increased both hepatic very low density lipoprotein (VLDL)-TG secretion and hepatic lipoprotein remnant clearance, associated a ~25% reduction in plasma TG levels. Conversely, reducing endogenous ApoF expression reduced VLDL secretion in vivo , and reduced hepatocyte VLDL uptake by ~15% in vitro . Transcriptomic analysis of APOF -overexpressing mouse livers revealed a gene signature related to enhanced ApoB-lipoprotein clearance, including increased expression of Ldlr and Lrp1 , among others. CONCLUSION These data reveal a previously undescribed role for ApoF in the control of plasma and hepatic lipoprotein metabolism by favoring VLDL-TG secretion and hepatic lipoprotein remnant particle clearance.
Collapse
Affiliation(s)
- Audrey Deprince
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011‐ EGID, Lille, France
| | - Nathalie Hennuyer
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011‐ EGID, Lille, France
| | - Sander Kooijman
- Division of Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Department of Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Amanda C. M. Pronk
- Division of Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Department of Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Eric Baugé
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011‐ EGID, Lille, France
| | - Viktor Lienard
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011‐ EGID, Lille, France
| | - An Verrijken
- Department of Endocrinology, Diabetology and Metabolism, Antwerp University Hospital, Antwerp, Belgium
- Laboratory of Experimental Medicine and Paediatrics, University of Antwerp, Antwerp, Belgium
| | - Eveline Dirinck
- Department of Endocrinology, Diabetology and Metabolism, Antwerp University Hospital, Antwerp, Belgium
- Laboratory of Experimental Medicine and Paediatrics, University of Antwerp, Antwerp, Belgium
| | - Luisa Vonghia
- Department of Gastroenterology Hepatology, Antwerp University Hospital, Antwerp, Belgium
- Laboratory of Experimental Medicine and Paediatrics, University of Antwerp, Antwerp, Belgium
| | - Eloïse Woitrain
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011‐ EGID, Lille, France
| | - Niels J. Kloosterhuis
- Department of Paediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Eléonore Marez
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011‐ EGID, Lille, France
| | - Pauline Jacquemain
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011‐ EGID, Lille, France
| | - Justina C. Wolters
- Department of Paediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Fanny Lalloyer
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011‐ EGID, Lille, France
| | - Delphine Eberlé
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011‐ EGID, Lille, France
| | - Sandrine Quemener
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011‐ EGID, Lille, France
| | - Emmanuelle Vallez
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011‐ EGID, Lille, France
| | - Anne Tailleux
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011‐ EGID, Lille, France
| | - Mostafa Kouach
- Univ. Lille, CHU Lille, ULR 7365‐GRITA‐Groupe de Recherche sur les formes Injectables et les Technologies Associées, Lille, France
| | - Jean‐Francois Goossens
- Univ. Lille, CHU Lille, ULR 7365‐GRITA‐Groupe de Recherche sur les formes Injectables et les Technologies Associées, Lille, France
| | - Violeta Raverdy
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1190 ‐ EGID, Lille, France
| | - Bruno Derudas
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011‐ EGID, Lille, France
| | - Jan Albert Kuivenhoven
- Department of Paediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Mikaël Croyal
- Université de Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France
- Université de Nantes, CHU Nantes, Inserm, CNRS, SFR Santé, Inserm UMS 016, CNRS UMS 3556, Nantes, France
- CRNH‐Ouest Mass Spectrometry Core Facility, Nantes, France
| | - Bart van de Sluis
- Department of Paediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Sven Francque
- Department of Gastroenterology Hepatology, Antwerp University Hospital, Antwerp, Belgium
- Laboratory of Experimental Medicine and Paediatrics, University of Antwerp, Antwerp, Belgium
| | - François Pattou
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1190 ‐ EGID, Lille, France
| | - Patrick C. N. Rensen
- Division of Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Department of Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Bart Staels
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011‐ EGID, Lille, France
| | - Joel T. Haas
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011‐ EGID, Lille, France
| |
Collapse
|
10
|
Afonso MB, Islam T, Magusto J, Amorim R, Lenoir V, Simões RF, Teixeira J, Silva LC, Wendum D, Jéru I, Vigouroux C, Castro RE, Oliveira PJ, Prip‐Buus C, Ratziu V, Gautheron J, Rodrigues CMP. RIPK3 dampens mitochondrial bioenergetics and lipid droplet dynamics in metabolic liver disease. Hepatology 2023; 77:1319-1334. [PMID: 36029129 PMCID: PMC10026966 DOI: 10.1002/hep.32756] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 08/16/2022] [Accepted: 08/17/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND AIMS Receptor-interacting protein kinase 3 (RIPK3) mediates NAFLD progression, but its metabolic function is unclear. Here, we aimed to investigate the role of RIPK3 in modulating mitochondria function, coupled with lipid droplet (LD) architecture in NAFLD. APPROACH AND RESULTS Functional studies evaluating mitochondria and LD biology were performed in wild-type (WT) and Ripk3-/- mice fed a choline-deficient, amino acid-defined (CDAA) diet for 32 and 66 weeks and in CRISPR-Cas9 Ripk3 -null fat-loaded immortalized hepatocytes. The association between hepatic perilipin (PLIN) 1 and 5, RIPK3, and disease severity was also addressed in a cohort of patients with NAFLD and in PLIN1 -associated familial partial lipodystrophy. Ripk3 deficiency rescued impairment in mitochondrial biogenesis, bioenergetics, and function in CDAA diet-fed mice and fat-loaded hepatocytes. Ripk3 deficiency was accompanied by a strong upregulation of antioxidant systems, leading to diminished oxidative stress upon fat loading both in vivo and in vitro. Strikingly, Ripk3-/- hepatocytes displayed smaller size LD in higher numbers than WT cells after incubation with free fatty acids. Ripk3 deficiency upregulated adipocyte and hepatic levels of LD-associated proteins PLIN1 and PLIN5. PLIN1 upregulation controlled LD structure and diminished mitochondrial stress upon free fatty acid overload in Ripk3-/- hepatocytes and was associated with diminished human NAFLD severity. Conversely, a pathogenic PLIN1 frameshift variant was associated with NAFLD and fibrosis, as well as with increased hepatic RIPK3 levels in familial partial lipodystrophy. CONCLUSIONS Ripk3 deficiency restores mitochondria bioenergetics and impacts LD dynamics. RIPK3 inhibition is promising in ameliorating NAFLD.
Collapse
Affiliation(s)
- Marta B. Afonso
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Tawhidul Islam
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Julie Magusto
- Institute of Cardiometabolism and Nutrition, Paris, France
- Sorbonne Université, Inserm, Centre de Recherche Saint‐Antoine, Paris, France
| | - Ricardo Amorim
- CNC ‐ Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Véronique Lenoir
- Université de Paris, Institut Cochin, INSERM, CNRS, Paris, France
| | - Rui F. Simões
- CNC ‐ Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - José Teixeira
- CNC ‐ Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Liana C. Silva
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Dominique Wendum
- Sorbonne Université, Inserm, Centre de Recherche Saint‐Antoine, Paris, France
- Service d'Anatomo‐Pathologie, Service d'Hépatologie, Centre de Référence Maladie Rare Maladies Inflammatoires des Voies Biliaires‐Hépatites Auto‐immunes, Paris, France
| | - Isabelle Jéru
- Institute of Cardiometabolism and Nutrition, Paris, France
- Sorbonne Université, Inserm, Centre de Recherche Saint‐Antoine, Paris, France
- Laboratoire commun de Biologie et Génétique Moléculaires, Hôpital Saint‐Antoine, Paris, France
| | - Corinne Vigouroux
- Institute of Cardiometabolism and Nutrition, Paris, France
- Sorbonne Université, Inserm, Centre de Recherche Saint‐Antoine, Paris, France
- Centre National de Référence des Pathologies Rares de l'Insulino‐Sécrétion et de l'Insulino‐Sensibilité, Service de Diabétologie et Endocrinologie de la Reproduction, Hôpital Saint‐Antoine, Paris, France
| | - Rui E. Castro
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Paulo J. Oliveira
- CNC ‐ Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Carina Prip‐Buus
- Université de Paris, Institut Cochin, INSERM, CNRS, Paris, France
| | - Vlad Ratziu
- Institute of Cardiometabolism and Nutrition, Paris, France
- Department of Hepatology, Assistance Publique‐Hôpitaux de Paris, Pitié‐Salpêtrière Hospital, Paris, France
- Sorbonne Université, Inserm, Centre de Recherche des Cordeliers, and ICAN Institute of Cardiometabolism and Nutrition, Paris, France
| | - Jérémie Gautheron
- Institute of Cardiometabolism and Nutrition, Paris, France
- Sorbonne Université, Inserm, Centre de Recherche Saint‐Antoine, Paris, France
| | - Cecília M. P. Rodrigues
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
11
|
Heng H, Li D, Su W, Liu X, Yu D, Bian Z, Li J. Exploration of comorbidity mechanisms and potential therapeutic targets of rheumatoid arthritis and pigmented villonodular synovitis using machine learning and bioinformatics analysis. Front Genet 2023; 13:1095058. [PMID: 36685864 PMCID: PMC9853060 DOI: 10.3389/fgene.2022.1095058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 12/21/2022] [Indexed: 01/08/2023] Open
Abstract
Background: Rheumatoid arthritis (RA) is a chronic autoimmune disease. Pigmented villonodular synovitis (PVNS) is a tenosynovial giant cell tumor that can involve joints. The mechanisms of co-morbidity between the two diseases have not been thoroughly explored. Therefore, this study focused on investigating the functions, immunological differences, and potential therapeutic targets of common genes between RA and PVNS. Methods: Through the dataset GSE3698 obtained from the Gene Expression Omnibus (GEO) database, the differentially expressed genes (DEGs) were screened by R software, and weighted gene coexpression network analysis (WGCNA) was performed to discover the modules most relevant to the clinical features. The common genes between the two diseases were identified. The molecular functions and biological processes of the common genes were analyzed. The protein-protein interaction (PPI) network was constructed using the STRING database, and the results were visualized in Cytoscape software. Two machine learning algorithms, least absolute shrinkage and selection operator (LASSO) logistic regression and random forest (RF) were utilized to identify hub genes and predict the diagnostic efficiency of hub genes as well as the correlation between immune infiltrating cells. Results: We obtained a total of 107 DEGs, a module (containing 250 genes) with the highest correlation with clinical characteristics, and 36 common genes after taking the intersection. Moreover, using two machine learning algorithms, we identified three hub genes (PLIN, PPAP2A, and TYROBP) between RA and PVNS and demonstrated good diagnostic performance using ROC curve and nomogram plots. Single sample Gene Set Enrichment Analysis (ssGSEA) was used to analyze the biological functions in which three genes were mostly engaged. Finally, three hub genes showed a substantial association with 28 immune infiltrating cells. Conclusion: PLIN, PPAP2A, and TYROBP may influence RA and PVNS by modulating immunity and contribute to the diagnosis and therapy of the two diseases.
Collapse
Affiliation(s)
- Hongquan Heng
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Dazhuang Li
- Department of Orthopedics, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Wenxing Su
- Department of Plastic and Burn Surgery, The Second Affiliated Hospital of Chengdu Medical College (China National Nuclear Corporation 416 Hospital), Chengdu, China
| | - Xinyue Liu
- Department of Radiology, Wangjiang Hospital of Sichuan University, Chengdu, China
| | - Daojiang Yu
- Department of Plastic and Burn Surgery, The Second Affiliated Hospital of Chengdu Medical College (China National Nuclear Corporation 416 Hospital), Chengdu, China,*Correspondence: Daojiang Yu, ; Zhengjun Bian, ; Jian Li,
| | - Zhengjun Bian
- Department of Orthopedics, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China,*Correspondence: Daojiang Yu, ; Zhengjun Bian, ; Jian Li,
| | - Jian Li
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China,*Correspondence: Daojiang Yu, ; Zhengjun Bian, ; Jian Li,
| |
Collapse
|
12
|
PNPLA3(I148M) Inhibits Lipolysis by Perilipin-5-Dependent Competition with ATGL. Cells 2022; 12:cells12010073. [PMID: 36611868 PMCID: PMC9818421 DOI: 10.3390/cells12010073] [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: 12/01/2022] [Revised: 12/19/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022] Open
Abstract
The single nucleotide polymorphism I148M of the lipase patatin-like phospholipase domain containing 3 (PNPLA3) is associated with an unfavorable prognosis in alcoholic and non-alcoholic steatohepatitis (ASH, NASH), with progression to liver cirrhosis and development of hepatocellular carcinoma. In this study, we investigated the mechanistic interaction of PNPLA3 with lipid droplet (LD)-associated proteins of the perilipin family, which serve as gatekeepers for LD degradation. In a collective of 106 NASH, ASH and control liver samples, immunohistochemical analyses revealed increased ballooning, inflammation and fibrosis, as well as an accumulation of PNPLA3-perilipin 5 complexes on larger LDs in patients homo- and heterozygous for PNPLA3(I148M). Co-immunoprecipitation demonstrated an interaction of PNPLA3 with perilipin 5 and the key enzyme of lipolysis, adipose triglyceride lipase (ATGL). Localization studies in cell cultures and human liver showed colocalization of perilipin 5, ATGL and PNPLA3. Moreover, the lipolytic activity of ATGL was negatively regulated by PNPLA3 and perilipin 5, whereas perilipin 1 displaced PNPLA3 from the ATGL complex. Furthermore, ballooned hepatocytes, the hallmark of steatohepatitis, were positive for PNPLA3 and perilipins 2 and 5, but showed decreased perilipin 1 expression with respect to neighboured hepatocytes. In summary, PNPLA3- and ATGL-driven lipolysis is significantly regulated by perilipin 1 and 5 in steatohepatitis.
Collapse
|
13
|
Schelbert S, Schindeldecker M, Drebber U, Witzel HR, Weinmann A, Dries V, Schirmacher P, Roth W, Straub BK. Lipid Droplet-Associated Proteins Perilipin 1 and 2: Molecular Markers of Steatosis and Microvesicular Steatotic Foci in Chronic Hepatitis C. Int J Mol Sci 2022; 23:ijms232415456. [PMID: 36555099 PMCID: PMC9778710 DOI: 10.3390/ijms232415456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/29/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
Chronic infection with hepatitis C (HCV) is a major risk factor in the development of cirrhosis and hepatocellular carcinoma. Lipid metabolism plays a major role in the replication and deposition of HCV at lipid droplets (LDs). We have demonstrated the importance of LD-associated proteins of the perilipin family in steatotic liver diseases. Using a large collection of 231 human liver biopsies with HCV, perilipins 1 and 2 have been localized to LDs of hepatocytes that correlate with the degree of steatosis and specific HCV genotypes, but not significantly with the HCV viral load. Perilipin 1- and 2-positive microvesicular steatotic foci were observed in 36% of HCV liver biopsies, and also in chronic hepatitis B, autoimmune hepatitis and mildly steatotic or normal livers, but less or none were observed in normal livers of younger patients. Microvesicular steatotic foci did not frequently overlap with glycogenotic/clear cell foci as determined by PAS stain in serial sections. Steatotic foci were detected in all liver zones with slight architectural disarrays, as demonstrated by immunohistochemical glutamine synthetase staining of zone three, but without elevated Ki67-proliferation rates. In conclusion, microvesicular steatotic foci are frequently found in chronic viral hepatitis, but the clinical significance of these foci is so far not clear.
Collapse
Affiliation(s)
- Selina Schelbert
- Institute of Pathology, University Medical Center Mainz, 55131 Mainz, Germany
- Institute of Pathology, University Hospital Wuerzburg, 97080 Wuerzburg, Germany
| | | | - Uta Drebber
- Institute of Pathology, University Clinic Cologne, 50931 Cologne, Germany
| | - Hagen Roland Witzel
- Institute of Pathology, University Medical Center Mainz, 55131 Mainz, Germany
| | - Arndt Weinmann
- Department of Internal Medicine, University Medical Center, 55131 Mainz, Germany
| | - Volker Dries
- Institute of Pathology, University Clinic Cologne, 50931 Cologne, Germany
| | - Peter Schirmacher
- Institute of Pathology, University Medical Center Heidelberg, 69120 Heidelberg, Germany
| | - Wilfried Roth
- Institute of Pathology, University Medical Center Mainz, 55131 Mainz, Germany
| | - Beate Katharina Straub
- Institute of Pathology, University Medical Center Mainz, 55131 Mainz, Germany
- Correspondence: ; Tel.: +49-6131-17-7307
| |
Collapse
|
14
|
Fu Y, Zhou Y, Shen L, Li X, Zhang H, Cui Y, Zhang K, Li W, Chen WD, Zhao S, Li Y, Ye W. Diagnostic and therapeutic strategies for non-alcoholic fatty liver disease. Front Pharmacol 2022; 13:973366. [PMID: 36408234 PMCID: PMC9666875 DOI: 10.3389/fphar.2022.973366] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 10/18/2022] [Indexed: 11/07/2022] Open
Abstract
The global incidence rate of non-alcoholic fatty liver disease (NAFLD) is approximately 25%. With the global increase in obesity and its associated metabolic syndromes, NAFLD has become an important cause of chronic liver disease in many countries. Despite recent advances in pathogenesis, diagnosis, and therapeutics, there are still challenges in its treatment. In this review, we briefly describe diagnostic methods, therapeutic targets, and drugs related to NAFLD. In particular, we focus on evaluating carbohydrate and lipid metabolism, lipotoxicity, cell death, inflammation, and fibrosis as potential therapeutic targets for NAFLD. We also summarized the clinical research progress in terms of drug development and combination therapy, thereby providing references for NAFLD drug development.
Collapse
Affiliation(s)
- Yajie Fu
- Key Laboratory of Receptors-Mediated Gene Regulation, Hebi Key Laboratory of Liver Disease, School of Basic Medical Sciences, The People’s Hospital of Hebi, Henan University, Kaifeng, China
| | - Yanzhi Zhou
- Key Laboratory of Receptors-Mediated Gene Regulation, Hebi Key Laboratory of Liver Disease, School of Basic Medical Sciences, The People’s Hospital of Hebi, Henan University, Kaifeng, China
| | - Linhu Shen
- Key Laboratory of Receptors-Mediated Gene Regulation, Hebi Key Laboratory of Liver Disease, School of Basic Medical Sciences, The People’s Hospital of Hebi, Henan University, Kaifeng, China
| | - Xuewen Li
- Key Laboratory of Receptors-Mediated Gene Regulation, Hebi Key Laboratory of Liver Disease, School of Basic Medical Sciences, The People’s Hospital of Hebi, Henan University, Kaifeng, China
| | - Haorui Zhang
- Key Laboratory of Receptors-Mediated Gene Regulation, Hebi Key Laboratory of Liver Disease, School of Basic Medical Sciences, The People’s Hospital of Hebi, Henan University, Kaifeng, China
| | - Yeqi Cui
- Key Laboratory of Receptors-Mediated Gene Regulation, Hebi Key Laboratory of Liver Disease, School of Basic Medical Sciences, The People’s Hospital of Hebi, Henan University, Kaifeng, China
| | - Ke Zhang
- Key Laboratory of Receptors-Mediated Gene Regulation, Hebi Key Laboratory of Liver Disease, School of Basic Medical Sciences, The People’s Hospital of Hebi, Henan University, Kaifeng, China
| | - Weiguo Li
- Key Laboratory of Receptors-Mediated Gene Regulation, Hebi Key Laboratory of Liver Disease, School of Basic Medical Sciences, The People’s Hospital of Hebi, Henan University, Kaifeng, China
| | - Wei-dong Chen
- Key Laboratory of Receptors-Mediated Gene Regulation, Hebi Key Laboratory of Liver Disease, School of Basic Medical Sciences, The People’s Hospital of Hebi, Henan University, Kaifeng, China
- Key Laboratory of Receptors-Mediated Gene Regulation and Drug Discovery, School of Basic Medical Science, Inner Mongolia Medical University, Hohhot, China
| | - Shizhen Zhao
- Key Laboratory of Receptors-Mediated Gene Regulation, Hebi Key Laboratory of Liver Disease, School of Basic Medical Sciences, The People’s Hospital of Hebi, Henan University, Kaifeng, China
- *Correspondence: Shizhen Zhao, ; Yunfu Li, ; Wenling Ye,
| | - Yunfu Li
- Key Laboratory of Receptors-Mediated Gene Regulation, Hebi Key Laboratory of Liver Disease, School of Basic Medical Sciences, The People’s Hospital of Hebi, Henan University, Kaifeng, China
- *Correspondence: Shizhen Zhao, ; Yunfu Li, ; Wenling Ye,
| | - Wenling Ye
- Key Laboratory of Receptors-Mediated Gene Regulation, Hebi Key Laboratory of Liver Disease, School of Basic Medical Sciences, The People’s Hospital of Hebi, Henan University, Kaifeng, China
- *Correspondence: Shizhen Zhao, ; Yunfu Li, ; Wenling Ye,
| |
Collapse
|
15
|
Su W, Wu S, Yang Y, Guo Y, Zhang H, Su J, Chen L, Mao Z, Lan R, Cao R, Wang C, Xu H, Zhang C, Li S, Gao M, Chen X, Zheng Z, Wang B, Liu Y, Liu Z, Wang Z, Liu B, Fan X, Zhang X, Guan Y. Phosphorylation of 17β-hydroxysteroid dehydrogenase 13 at serine 33 attenuates nonalcoholic fatty liver disease in mice. Nat Commun 2022; 13:6577. [PMID: 36323699 PMCID: PMC9630536 DOI: 10.1038/s41467-022-34299-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 10/20/2022] [Indexed: 11/06/2022] Open
Abstract
17β-hydroxysteroid dehydrogenase-13 is a hepatocyte-specific, lipid droplet-associated protein. A common loss-of-function variant of HSD17B13 (rs72613567: TA) protects patients against non-alcoholic fatty liver disease with underlying mechanism incompletely understood. In the present study, we identify the serine 33 of 17β-HSD13 as an evolutionally conserved PKA target site and its phosphorylation facilitates lipolysis by promoting its interaction with ATGL on lipid droplets. Targeted mutation of Ser33 to Ala (S33A) decreases ATGL-dependent lipolysis in cultured hepatocytes by reducing CGI-58-mediated ATGL activation. Importantly, a transgenic knock-in mouse strain carrying the HSD17B13 S33A mutation (HSD17B1333A/A) spontaneously develops hepatic steatosis with reduced lipolysis and increased inflammation. Moreover, Hsd17B1333A/A mice are more susceptible to high-fat diet-induced nonalcoholic steatohepatitis. Finally, we find reproterol, a potential 17β-HSD13 modulator and FDA-approved drug, confers a protection against nonalcoholic steatohepatitis via PKA-mediated Ser33 phosphorylation of 17β-HSD13. Therefore, targeting the Ser33 phosphorylation site could represent a potential approach to treat NASH.
Collapse
Affiliation(s)
- Wen Su
- grid.263488.30000 0001 0472 9649Department of Pathophysiology, Shenzhen University, Shenzhen, 518060 China ,Shenzhen University Health Science Center, Shenzhen University, Shenzhen, 518060 China
| | - Sijin Wu
- grid.9227.e0000000119573309State Key Laboratory of Molecular Reaction Dynamics, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116024 China
| | - Yongliang Yang
- grid.30055.330000 0000 9247 7930Laboratoy of Innovative Drug Discovery, School of Bioengineering, Dalian University of Technology, Dalian, 116023 China
| | - Yanlin Guo
- grid.22069.3f0000 0004 0369 6365Health Science Center, East China Normal University, Shanghai, 200241 China
| | - Haibo Zhang
- grid.411971.b0000 0000 9558 1426Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044 China
| | - Jie Su
- grid.263488.30000 0001 0472 9649Department of Pathophysiology, Shenzhen University, Shenzhen, 518060 China ,Shenzhen University Health Science Center, Shenzhen University, Shenzhen, 518060 China
| | - Lei Chen
- grid.263488.30000 0001 0472 9649Department of Pathophysiology, Shenzhen University, Shenzhen, 518060 China ,Shenzhen University Health Science Center, Shenzhen University, Shenzhen, 518060 China
| | - Zhuo Mao
- Shenzhen University Health Science Center, Shenzhen University, Shenzhen, 518060 China
| | - Rongfeng Lan
- Shenzhen University Health Science Center, Shenzhen University, Shenzhen, 518060 China
| | - Rong Cao
- grid.263488.30000 0001 0472 9649Department of Nephrology, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035 China
| | - Chunjiong Wang
- grid.265021.20000 0000 9792 1228Department of Physiology and Pathophysiology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, Tianjin, China
| | - Hu Xu
- grid.411971.b0000 0000 9558 1426Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044 China
| | - Cong Zhang
- grid.411971.b0000 0000 9558 1426Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044 China
| | - Sha Li
- grid.412028.d0000 0004 1757 5708Medical College, Hebei University of Engineering, Handan, China
| | - Min Gao
- Shenzhen University Health Science Center, Shenzhen University, Shenzhen, 518060 China
| | - Xiaocong Chen
- Shenzhen University Health Science Center, Shenzhen University, Shenzhen, 518060 China
| | - Zhiyou Zheng
- Shenzhen University Health Science Center, Shenzhen University, Shenzhen, 518060 China
| | - Bing Wang
- grid.411971.b0000 0000 9558 1426Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044 China
| | - Yi’ao Liu
- Shenzhen University Health Science Center, Shenzhen University, Shenzhen, 518060 China
| | - Zuojun Liu
- Shenzhen University Health Science Center, Shenzhen University, Shenzhen, 518060 China
| | - Zimei Wang
- Shenzhen University Health Science Center, Shenzhen University, Shenzhen, 518060 China
| | - Baohua Liu
- Shenzhen University Health Science Center, Shenzhen University, Shenzhen, 518060 China
| | - Xinmin Fan
- grid.263488.30000 0001 0472 9649Department of Pathophysiology, Shenzhen University, Shenzhen, 518060 China ,Shenzhen University Health Science Center, Shenzhen University, Shenzhen, 518060 China
| | - Xiaoyan Zhang
- grid.22069.3f0000 0004 0369 6365Health Science Center, East China Normal University, Shanghai, 200241 China
| | - Youfei Guan
- grid.411971.b0000 0000 9558 1426Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044 China ,grid.411971.b0000 0000 9558 1426Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Dalian Medical University, Dalian, 116044 China
| |
Collapse
|
16
|
Gabbia D, Roverso M, Zanotto I, Colognesi M, Sayaf K, Sarcognato S, Arcidiacono D, Zaramella A, Realdon S, Ferri N, Guido M, Russo FP, Bogialli S, Carrara M, De Martin S. A Nutraceutical Formulation Containing Brown Algae Reduces Hepatic Lipid Accumulation by Modulating Lipid Metabolism and Inflammation in Experimental Models of NAFLD and NASH. Mar Drugs 2022; 20:572. [PMID: 36135761 PMCID: PMC9501409 DOI: 10.3390/md20090572] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 08/30/2022] [Accepted: 09/05/2022] [Indexed: 01/08/2023] Open
Abstract
Recently, some preclinical and clinical studies have demonstrated the ability of brown seaweeds in reducing the risk factors for metabolic syndrome. Here, we analyzed the beneficial effect of a nutraceutical formulation containing a phytocomplex extracted from seaweeds and chromium picolinate in animal models of liver steatosis of differing severities (rats with non-alcoholic fatty liver disease (NAFLD) and its complication, non-alcoholic steatohepatitis (NASH)). This treatment led to a significant drop in hepatic fat deposition in both models (p < 0.01 vs. untreated animals), accompanied by a reduction in plasma inflammatory cytokines, such as interleukin 6, tumor necrosis factor α, and C reactive protein, and myeloperoxidase expression in liver tissue. Furthermore, a modulation of the molecular pathways involved in lipid metabolism and storage was demonstrated, since we observed the significant reduction of the mRNA levels of fatty acid synthase, diacylglycerol acyltransferases, the sterol-binding protein SREBP-1, and the lipid transporter perilipin-2, in both treated NAFLD and NASH rats in comparison to untreated ones. In conclusion, this nutraceutical product was effective in reducing liver steatosis and showed further beneficial effects on hepatic inflammation and glycemic control, which were particularly evident in rats characterized by a more severe condition, thus representing a therapeutic option for the treatment of NAFLD and NASH patients.
Collapse
Affiliation(s)
- Daniela Gabbia
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy
| | - Marco Roverso
- Department of Chemical Sciences, University of Padova, 35131 Padova, Italy
| | - Ilaria Zanotto
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy
| | - Martina Colognesi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy
| | - Katia Sayaf
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35131 Padova, Italy
| | - Samantha Sarcognato
- Department of Pathology, Azienda ULSS2 Marca Trevigiana, 31100 Treviso, Italy
| | - Diletta Arcidiacono
- Gastroenterology Unit, Veneto Institute of Oncology IOV-IRCCS, 35131 Padova, Italy
| | - Alice Zaramella
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35131 Padova, Italy
- Gastroenterology Unit, Veneto Institute of Oncology IOV-IRCCS, 35131 Padova, Italy
| | - Stefano Realdon
- Gastroenterology Unit, Veneto Institute of Oncology IOV-IRCCS, 35131 Padova, Italy
| | - Nicola Ferri
- Department of Medicine, University of Padova, 35131 Padova, Italy
| | - Maria Guido
- Department of Pathology, Azienda ULSS2 Marca Trevigiana, 31100 Treviso, Italy
- Department of Medicine, University of Padova, 35131 Padova, Italy
| | - Francesco Paolo Russo
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35131 Padova, Italy
| | - Sara Bogialli
- Department of Chemical Sciences, University of Padova, 35131 Padova, Italy
| | - Maria Carrara
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy
| | - Sara De Martin
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy
| |
Collapse
|
17
|
Siemienowicz KJ, Filis P, Thomas J, Fowler PA, Duncan WC, Rae MT. Hepatic Mitochondrial Dysfunction and Risk of Liver Disease in an Ovine Model of “PCOS Males”. Biomedicines 2022; 10:biomedicines10061291. [PMID: 35740312 PMCID: PMC9220073 DOI: 10.3390/biomedicines10061291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 05/25/2022] [Accepted: 05/27/2022] [Indexed: 12/04/2022] Open
Abstract
First-degree male relatives of polycystic ovary syndrome (PCOS) sufferers can develop metabolic abnormalities evidenced by elevated circulating cholesterol and triglycerides, suggestive of a male PCOS equivalent. Similarly, male sheep overexposed to excess androgens in fetal life develop dyslipidaemia in adolescence. Dyslipidaemia, altered lipid metabolism, and dysfunctional hepatic mitochondria are associated with the development of non-alcoholic liver disease (NAFLD). We therefore dissected hepatic mitochondrial function and lipid metabolism in adolescent prenatally androgenized (PA) males from an ovine model of PCOS. Testosterone was directly administered to male ovine fetuses to create prenatal androgenic overexposure. Liver RNA sequencing and proteomics occurred at 6 months of age. Hepatic lipids, glycogen, ATP, reactive oxygen species (ROS), DNA damage, and collagen were assessed. Adolescent PA males had an increased accumulation of hepatic cholesterol and glycogen, together with perturbed glucose and fatty acid metabolism, mitochondrial dysfunction, with altered mitochondrial transport, decreased oxidative phosphorylation and ATP synthesis, and impaired mitophagy. Mitochondrial dysfunction in PA males was associated with increased hepatic ROS level and signs of early liver fibrosis, with clinical relevance to NAFLD progression. We conclude that excess in utero androgen exposure in male fetuses leads to a PCOS-like metabolic phenotype with dysregulated mitochondrial function and likely lifelong health sequelae.
Collapse
Affiliation(s)
- Katarzyna J. Siemienowicz
- School of Applied Science, Edinburgh Napier University, Edinburgh EH11 4BN, UK; (J.T.); (M.T.R.)
- MRC Centre for Reproductive Health, The University of Edinburgh, Edinburgh EH16 4TJ, UK;
- Correspondence:
| | - Panagiotis Filis
- Institute of Medical Sciences, School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK; (P.F.); (P.A.F.)
| | - Jennifer Thomas
- School of Applied Science, Edinburgh Napier University, Edinburgh EH11 4BN, UK; (J.T.); (M.T.R.)
| | - Paul A. Fowler
- Institute of Medical Sciences, School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK; (P.F.); (P.A.F.)
| | - W. Colin Duncan
- MRC Centre for Reproductive Health, The University of Edinburgh, Edinburgh EH16 4TJ, UK;
| | - Mick T. Rae
- School of Applied Science, Edinburgh Napier University, Edinburgh EH11 4BN, UK; (J.T.); (M.T.R.)
| |
Collapse
|
18
|
Takemura M, Mochizuki K, Harada Y, Okajima A, Hayakawa M, Dai P, Itoh Y, Tanaka H. Label-free Assessment of the Nascent State of Rat Non-alcoholic Fatty Liver Disease Using Spontaneous Raman Microscopy. Acta Histochem Cytochem 2022; 55:57-66. [PMID: 35509867 PMCID: PMC9043435 DOI: 10.1267/ahc.22-00013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 02/08/2022] [Indexed: 11/22/2022] Open
Abstract
Spontaneous Raman microscopy, which can detect molecular vibrations in cells and tissues, could be a useful tool for the label-free assessment of non-alcoholic fatty liver disease (NAFLD). However, it is unclear whether it can be used to evaluate the nascent state of NAFLD. To address this, we analyzed the Raman spectra of rat liver tissues in the nascent state of NAFLD upon excitation at 532 nm. Raman and histochemical analyses were performed of liver tissues from rats fed a high-fat, high-cholesterol diet (HFHCD). Raman microscopic imaging analysis of formalin-fixed thin tissue slices showed hepatic steatosis, as revealed by the Raman band at 2,854 cm-1, whereas lipid droplets were not detectable by hematoxylin-eosin staining of images until 3 days after feeding a HFHCD. Raman signals of retinol at 1,588 cm-1 emitted from hepatic stellate cells were distributed alongside hepatic cords; the retinol content rapidly decreased after feeding a HFHCD, whereas hepatic lipid content increased inversely. Raman microscopic analysis of the surface of fresh ex vivo livers enabled early detection of lipid accumulation after a 1-day feeding a HFHCD. In conclusion, spontaneous Raman microscopy can be applied to the label-free evaluation of the nascent state of NAFLD liver tissues.
Collapse
Affiliation(s)
- Masashi Takemura
- Department of Pathology and Cell Regulation, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho Kamigyo-ku, Kyoto 602–8566, Japan
- Department of Molecular Gatroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho Kamigyo-ku, Kyoto 602–8566, Japan
| | - Kentaro Mochizuki
- Department of Pathology and Cell Regulation, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho Kamigyo-ku, Kyoto 602–8566, Japan
| | - Yoshinori Harada
- Department of Pathology and Cell Regulation, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho Kamigyo-ku, Kyoto 602–8566, Japan
| | - Akira Okajima
- Department of Pathology and Cell Regulation, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho Kamigyo-ku, Kyoto 602–8566, Japan
- Department of Molecular Gatroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho Kamigyo-ku, Kyoto 602–8566, Japan
| | - Michiyo Hayakawa
- Department of Pathology and Cell Regulation, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho Kamigyo-ku, Kyoto 602–8566, Japan
| | - Ping Dai
- Department of Cellular Regenerative Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho Kamigyo-ku, Kyoto 602–8566, Japan
| | - Yoshito Itoh
- Department of Molecular Gatroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho Kamigyo-ku, Kyoto 602–8566, Japan
| | - Hideo Tanaka
- Department of Pathology and Cell Regulation, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho Kamigyo-ku, Kyoto 602–8566, Japan
| |
Collapse
|
19
|
Preventive Effects of Ilex Cornuta Aqueous Extract on High-Fat Diet-Induced Fatty Liver of Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:7183471. [PMID: 35432557 PMCID: PMC9010189 DOI: 10.1155/2022/7183471] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 03/09/2022] [Indexed: 12/26/2022]
Abstract
Objective To investigate the preventive effects of Ilex cornuta aqueous extract (ICAE) on high-fat diet (HFD)-induced fatty liver of mice and its mechanisms. Materials and Methods Twenty-six male KM (Kunming) mice were divided into 3 groups, including the control group (n = 9), fed with normal diet; HFD group (n = 9), fed with HFD; ICAE + HFD group (n = 8), fed with HFD and administered with ICAE (3 g·kg-1·d-1) at the same time for 10 weeks. Body weight, liver weight, intra-abdominal and subcutaneous fat weight, serum triglyceride (TG), total cholesterol (TC), and blood glucose were determined to evaluate the preventive effects of ICAE on obesity. The average 24 h food consumption of the mice was monitored for 5 times in the 9th week of the experiment to investigate the effects of ICAE on food intake. Serum alanine transaminase (ALT) and aspartate aminotransferase (AST) were assayed to observe the influences of HFD and ICAE on liver function. HE staining was adopted to observe the influence of ICAE on the morphology of adipose tissue and liver tissue. Hepatic TG and TC content assay and oil red O staining were used to evaluate the influences of ICAE on HFD-induced fatty liver, and the protein expression of peroxisome proliferator-activated receptors γ (PPARγ) and adipose differentiation-related protein (ADRP) in liver were examined by immunoblotting. Results ICAE treatment significantly reduced the increase of body weight, intra-abdominal, and subcutaneous fat and liver weight induced by HFD (P < 0.001), but has no influence on food intake; ICAE treatment attenuated the elevation of serum TG, TC, and glucose, as well as serum ALT and AST (P < 0.01, P < 0.05, P < 0.001) and dramatically decreased the content of TG in liver (P < 0.01), but has no influence on hepatic TC content. HE staining and oil red O staining showed that ICAE significantly reduced HFD-induced white adipocyte hypertrophy and significantly inhibited lipid accumulation in liver. Immunoblotting showed that the protein levels of PPARγ and ADRP were significantly increased by HFD induction, which can be dramatically reduced by ICAE treatment (P < 0.05, P < 0.0001). Conclusion ICAE has preventive effects on HFD-induced obesity and fatty liver in mice, exerted beneficial effects upon HFD-induced hepatic injury. The preventive effects of ICAE on fatty liver are concerned with the downregulation of PPARγ and ADRP protein expression in liver.
Collapse
|
20
|
Gong L, Wang GE, Ma QY, Hao WZ, Xian MH, Wu YP, Kurihara H, He RR, Chen JX. Novel insights into the effect of Xiaoyao san on corticosterone-induced hepatic steatosis: inhibition of glucocorticoid receptor/perilipin-2 signaling pathway. ACUPUNCTURE AND HERBAL MEDICINE 2022; 2:49-57. [DOI: 10.1097/hm9.0000000000000011] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 12/02/2021] [Indexed: 01/03/2025]
Abstract
Abstract
Objective:
Xiaoyao san (XYS) is a classic traditional Chinese medicinal formula. It has been clinically administered to regulate liver function. However, its mechanisms in glucocorticoid-induced hepatic steatosis are unknown. This study aimed to investigate whether XYS protects against corticosterone (CORT)-induced hepatic steatosis, and to explore its mechanism.
Methods:
High-fat diet mice induced with hepatic steatosis by 2 mg/kg CORT were administered 2.56 g/kg or 5.12 g/kg XYS daily for 7 weeks. The effects of XYS on hepatic steatosis in mice were evaluated by H&E and Oil Red O staining and by measuring their plasma lipids (triglyceride, total cholesterol, and free fatty acids). The mechanism of XYS against hepatic steatosis was investigated by network pharmacology, immunohistochemistry, western blotting, and gain-of-function/loss-of-function experiments.
Results:
XYS alleviated CORT-induced steatosis, decreased plasma lipids, and inhibited glucocorticoid receptor (GR) activation in the liver. Network pharmacology data indicated that XYS may have mitigated hepatic steatosis via GR which mediated adipose differentiation-related protein (ADFP). Gain-of-function/loss-of-function experiments in vitro confirmed that GR positively regulated ADFP expression.
Conclusions:
XYS ameliorated CORT-induced hepatic steatosis by downregulating the GR/ADFP axis and inhibiting lipid metabolism. Our studies implicate that XYS is promising as a therapy for CORT-induced hepatic steatosis, and lay the foundation for designing novel prophylactic and therapeutic strategies on CORT-induced hepatic steatosis.
Collapse
Affiliation(s)
- Lian Gong
- Formula-Pattern Research Center, School of Chinese Medicine, Jinan University, Guangzhou, China
| | - Guo-En Wang
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou, China
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Qing-Yu Ma
- Formula-Pattern Research Center, School of Chinese Medicine, Jinan University, Guangzhou, China
| | - Wen-Zhi Hao
- Formula-Pattern Research Center, School of Chinese Medicine, Jinan University, Guangzhou, China
| | - Min-Hua Xian
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yan-Ping Wu
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou, China
| | - Hiroshi Kurihara
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou, China
| | - Rong-Rong He
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou, China
| | - Jia-Xu Chen
- Formula-Pattern Research Center, School of Chinese Medicine, Jinan University, Guangzhou, China
| |
Collapse
|
21
|
The Bank Vole (Clethrionomys glareolus)—Small Animal Model for Hepacivirus Infection. Viruses 2021; 13:v13122421. [PMID: 34960690 PMCID: PMC8708279 DOI: 10.3390/v13122421] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 11/29/2021] [Indexed: 12/15/2022] Open
Abstract
Many people worldwide suffer from hepatitis C virus (HCV) infection, which is frequently persistent. The lack of efficient vaccines against HCV and the unavailability of or limited compliance with existing antiviral therapies is problematic for health care systems worldwide. Improved small animal models would support further hepacivirus research, including development of vaccines and novel antivirals. The recent discovery of several mammalian hepaciviruses may facilitate such research. In this study, we demonstrated that bank voles (Clethrionomys glareolus) were susceptible to bank vole-associated Hepacivirus F and Hepacivirus J strains, based on the detection of hepaciviral RNA in 52 of 55 experimentally inoculated voles. In contrast, interferon α/β receptor deficient C57/Bl6 mice were resistant to infection with both bank vole hepaciviruses (BvHVs). The highest viral genome loads in infected voles were detected in the liver, and viral RNA was visualized by in situ hybridization in hepatocytes, confirming a marked hepatotropism. Furthermore, liver lesions in infected voles resembled those of HCV infection in humans. In conclusion, infection with both BvHVs in their natural hosts shares striking similarities to HCV infection in humans and may represent promising small animal models for this important human disease.
Collapse
|
22
|
Garcia-Macia M, Santos-Ledo A, Leslie J, Paish HL, Collins AL, Scott RS, Watson A, Burgoyne RA, White S, French J, Hammond J, Borthwick LA, Mann J, Bolaños JP, Korolchuk VI, Oakley F, Mann DA. A Mammalian Target of Rapamycin-Perilipin 3 (mTORC1-Plin3) Pathway is essential to Activate Lipophagy and Protects Against Hepatosteatosis. Hepatology 2021; 74:3441-3459. [PMID: 34233024 DOI: 10.1002/hep.32048] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 05/28/2021] [Accepted: 06/13/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIMS NAFLD is the most common hepatic pathology in western countries and no treatment is currently available. NAFLD is characterized by the aberrant hepatocellular accumulation of fatty acids in the form of lipid droplets (LDs). Recently, it was shown that liver LD degradation occurs through a process termed lipophagy, a form of autophagy. However, the molecular mechanisms governing liver lipophagy are elusive. Here, we aimed to ascertain the key molecular players that regulate hepatic lipophagy and their importance in NAFLD. APPROACH AND RESULTS We analyzed the formation and degradation of LD in vitro (fibroblasts and primary mouse hepatocytes), in vivo and ex vivo (mouse and human liver slices) and focused on the role of the autophagy master regulator mammalian target of rapamycin complex (mTORC) 1 and the LD coating protein perilipin (Plin) 3 in these processes. We show that the autophagy machinery is recruited to the LD on hepatic overload of oleic acid in all experimental settings. This led to activation of lipophagy, a process that was abolished by Plin3 knockdown using RNA interference. Furthermore, Plin3 directly interacted with the autophagy proteins focal adhesion interaction protein 200 KDa and autophagy-related 16L, suggesting that Plin3 functions as a docking protein or is involved in autophagosome formation to activate lipophagy. Finally, we show that mTORC1 phosphorylated Plin3 to promote LD degradation. CONCLUSIONS These results reveal that mTORC1 regulates liver lipophagy through a mechanism dependent on Plin3 phosphorylation. We propose that stimulating this pathway can enhance lipophagy in hepatocytes to help protect the liver from lipid-mediated toxicity, thus offering a therapeutic strategy in NAFLD.
Collapse
Affiliation(s)
- Marina Garcia-Macia
- Newcastle Fibrosis Research Group, Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- Institute of Biomedical Research of Salamanca, University Hospital of Salamanca, Salamanca, Spain
- Institute of Functional Biology and Genomics, University of Salamanca, CSIC, Salamanca, Spain
- Centro de Investigación Biomédica en Red sobre Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Jack Leslie
- Newcastle Fibrosis Research Group, Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Hannah L Paish
- Newcastle Fibrosis Research Group, Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Amy L Collins
- Newcastle Fibrosis Research Group, Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Rebecca S Scott
- Newcastle Fibrosis Research Group, Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- FibroFind Ltd, William Leech Building, Medical School, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Abigail Watson
- FibroFind Ltd, William Leech Building, Medical School, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Rachel A Burgoyne
- Newcastle Fibrosis Research Group, Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Steve White
- Department of Hepatobiliary Surgery, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Jeremy French
- Department of Hepatobiliary Surgery, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - John Hammond
- Department of Hepatobiliary Surgery, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Lee A Borthwick
- Newcastle Fibrosis Research Group, Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Jelena Mann
- Newcastle Fibrosis Research Group, Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Juan P Bolaños
- Institute of Biomedical Research of Salamanca, University Hospital of Salamanca, Salamanca, Spain
- Institute of Functional Biology and Genomics, University of Salamanca, CSIC, Salamanca, Spain
- Centro de Investigación Biomédica en Red sobre Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Viktor I Korolchuk
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Fiona Oakley
- Newcastle Fibrosis Research Group, Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Derek A Mann
- Newcastle Fibrosis Research Group, Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
23
|
Kalyesubula M, Mopuri R, Asiku J, Rosov A, Yosefi S, Edery N, Bocobza S, Moallem U, Dvir H. High-dose vitamin B1 therapy prevents the development of experimental fatty liver driven by overnutrition. Dis Model Mech 2021; 14:dmm.048355. [PMID: 33608323 PMCID: PMC7988776 DOI: 10.1242/dmm.048355] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 01/11/2021] [Indexed: 12/16/2022] Open
Abstract
Fatty liver is an abnormal metabolic condition of excess intrahepatic fat. This condition, referred to as hepatic steatosis, is tightly associated with chronic liver disease and systemic metabolic morbidity. The most prevalent form in humans, i.e. non-alcoholic fatty liver, generally develops due to overnutrition and sedentary lifestyle, and has as yet no approved drug therapy. Previously, we have developed a relevant large-animal model in which overnourished sheep raised on a high-calorie carbohydrate-rich diet develop hyperglycemia, hyperinsulinemia, insulin resistance, and hepatic steatosis. Here, we tested the hypothesis that treatment with thiamine (vitamin B1) can counter the development of hepatic steatosis driven by overnutrition. Remarkably, the thiamine-treated animals presented with completely normal levels of intrahepatic fat, despite consuming the same amount of liver-fattening diet. Thiamine treatment also decreased hyperglycemia and increased the glycogen content of the liver, but it did not improve insulin sensitivity, suggesting that steatosis can be addressed independently of targeting insulin resistance. Thiamine increased the catalytic capacity for hepatic oxidation of carbohydrates and fatty acids. However, at gene-expression levels, more-pronounced effects were observed on lipid-droplet formation and lipidation of very-low-density lipoprotein, suggesting that thiamine affects lipid metabolism not only through its known classic coenzyme roles. This discovery of the potent anti-steatotic effect of thiamine may prove clinically useful in managing fatty liver-related disorders.This article has an associated First Person interview with the joint first authors of the paper.
Collapse
Affiliation(s)
- Mugagga Kalyesubula
- Institute of Animal Science, Volcani Center - Agricultural Research Organization (ARO), Rishon LeZion 7528809, Israel.,Department of Animal Science, The Hebrew University of Jerusalem, Rehovot 7610001, Israel
| | - Ramgopal Mopuri
- Institute of Animal Science, Volcani Center - Agricultural Research Organization (ARO), Rishon LeZion 7528809, Israel
| | - Jimmy Asiku
- Institute of Animal Science, Volcani Center - Agricultural Research Organization (ARO), Rishon LeZion 7528809, Israel.,Institute of Biochemistry, Food Science and Nutrition, The Hebrew University of Jerusalem, Rehovot 7610001, Israel
| | - Alexander Rosov
- Institute of Animal Science, Volcani Center - Agricultural Research Organization (ARO), Rishon LeZion 7528809, Israel
| | - Sara Yosefi
- Institute of Animal Science, Volcani Center - Agricultural Research Organization (ARO), Rishon LeZion 7528809, Israel
| | - Nir Edery
- Pathology Laboratory, Kimron Veterinary Institute, Veterinary Services, Rishon LeZion 50250, Israel
| | - Samuel Bocobza
- Institute of Plant Sciences, Volcani Center - ARO, Rishon LeZion 7528809, Israel
| | - Uzi Moallem
- Institute of Animal Science, Volcani Center - Agricultural Research Organization (ARO), Rishon LeZion 7528809, Israel
| | - Hay Dvir
- Institute of Animal Science, Volcani Center - Agricultural Research Organization (ARO), Rishon LeZion 7528809, Israel
| |
Collapse
|
24
|
Fowl Adenovirus Serotype 4 Induces Hepatic Steatosis via Activation of Liver X Receptor-α. J Virol 2021; 95:JVI.01938-20. [PMID: 33361420 DOI: 10.1128/jvi.01938-20] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 12/09/2020] [Indexed: 12/24/2022] Open
Abstract
Fowl adenovirus serotype 4 (FAdV-4) is a hepatotropic virus that causes severe hepatic damage characterized by basophilic intranuclear inclusion bodies, vacuolar degeneration, and multifocal necrosis in hepatocytes. Many aspects of FAdV-4 infection and pathogenesis, however, remain unknown. Here, we found that FAdV-4-induced hepatic injury is accompanied by the accumulation of oil droplets (triglycerides) in the cytoplasm of hepatocytes, a typical indicator of steatosis, in FAdV-4-infected chickens. Significant upregulation of adipose synthesis-related genes, such as liver X receptor-α (LXR-α), peroxisome proliferator-activated receptor gamma (PPAR-γ), and sterol regulatory element-binding protein-1c (SREBP-1c), and significant downregulation of low-density lipoprotein secretion-related genes and lipid oxidation- and lipid decomposition-related genes were observed in the infected chickens. FAdV-4 infection in cultured leghorn male hepatoma (LMH) cells caused similar signs of steatosis, with alterations in various lipogenesis-related genes. We eliminated the effect of LXR-α activation on FAdV-4-induced steatosis and found that treatment with an LXR-α antagonist (SR9243) and RNA interference (small interfering RNA targeting LXR-α [Si-LXR-α]) decreased the number of oil droplets and the accumulation of lipogenic genes, but treatment with an LXR-α agonist (T0901317) increased the number of oil droplets and the accumulation of lipogenic genes in the cells. Additionally, SR9243 treatment or Si-LXR-α transfection led to significant reductions in viral DNA level, protein expression, and virus production, whereas T0901317 treatment caused significant increases in viral DNA level, protein expression, and virus production. However, inhibition of SREBP-1c activity had no significant effect on virus production. Collectively, these results indicated that FAdV-4-induced steatosis involves activation of the LXR-α signaling pathway, which might be a molecular mechanism underlying the hepatic injury associated with FAdV-4 infection.IMPORTANCE Fowl adenovirus serotype 4 (FAdV-4) is an important hepatotropic adenovirus in chicken, but the underlying mechanism of FAdV-4-induced hepatic injury remains unclear. We report here that infection with FAdV-4 induced the accumulation of oil droplets (triglycerides) in the cytoplasm of hepatocytes, a typical indicator of steatosis, in the livers of chickens. FAdV-4-induced steatosis might be caused by a disrupted balance of fat metabolism, as evidenced by differential regulation of various lipase genes. The significant upregulation of liver X receptor-α (LXR-α) prompted us to investigate the interplay between LXR-α activation and FAdV-4-induced steatosis. Treatment with an agonist, an antagonist, or RNA interference targeting LXR-α in cultured leghorn male hepatoma (LMH) cells indicated that FAdV-4-induced steatosis was dependent upon LXR-α activation, which contributed to virus replication. These results provide important mechanistic insights, revealing that FAdV-4 induces hepatic steatosis by activating the LXR-α signaling pathway and highlighting the therapeutic potential of strategies targeting the LXR-α pathway for the treatment of FAdV-4 infection.
Collapse
|
25
|
Herker E, Vieyres G, Beller M, Krahmer N, Bohnert M. Lipid Droplet Contact Sites in Health and Disease. Trends Cell Biol 2021; 31:345-358. [PMID: 33546922 DOI: 10.1016/j.tcb.2021.01.004] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 01/05/2021] [Accepted: 01/08/2021] [Indexed: 01/04/2023]
Abstract
After having been disregarded for a long time as inert fat drops, lipid droplets (LDs) are now recognized as ubiquitous cellular organelles with key functions in lipid biology and beyond. The identification of abundant LD contact sites, places at which LDs are physically attached to other organelles, has uncovered an unexpected level of communication between LDs and the rest of the cell. In recent years, many disease factors mutated in hereditary disorders have been recognized as LD contact site proteins. Furthermore, LD contact sites are dramatically rearranged in response to infections with intracellular pathogens, as well as under pathological metabolic conditions such as hepatic steatosis. Collectively, it is emerging that LD-organelle contacts are important players in development and progression of disease.
Collapse
Affiliation(s)
- Eva Herker
- Institute of Virology, Philipps-University Marburg, 35043 Marburg, Germany.
| | - Gabrielle Vieyres
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany; Leibniz ScienceCampus InterACt, Hamburg, Germany.
| | - Mathias Beller
- Institute for Mathematical Modeling of Biological Systems, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; Systems Biology of Lipid Metabolism, Heinrich Heine University Düsseldorf, Germany.
| | - Natalie Krahmer
- Institute for Diabetes and Obesity, Helmholtz Zentrum München, 85764 Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany.
| | - Maria Bohnert
- Institute of Cell Dynamics and Imaging, University of Münster, 48149 Münster, Germany; Cells in Motion Interfaculty Centre (CiM), University of Münster, Münster, Germany.
| |
Collapse
|
26
|
Graffmann N, Ncube A, Martins S, Fiszl AR, Reuther P, Bohndorf M, Wruck W, Beller M, Czekelius C, Adjaye J. A stem cell based in vitro model of NAFLD enables the analysis of patient specific individual metabolic adaptations in response to a high fat diet and AdipoRon interference. Biol Open 2021; 10:bio.054189. [PMID: 33372064 PMCID: PMC7860118 DOI: 10.1242/bio.054189] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a multifactorial disease. Its development and progression depend on genetically predisposed susceptibility of the patient towards several ‘hits’ that induce fat storage first and later inflammation and fibrosis. Here, we differentiated induced pluripotent stem cells (iPSCs) derived from four distinct donors with varying disease stages into hepatocyte like cells (HLCs) and determined fat storage as well as metabolic adaptations after stimulations with oleic acid. We could recapitulate the complex networks that control lipid and glucose metabolism and we identified distinct gene expression profiles related to the steatosis phenotype of the donor. In an attempt to reverse the steatotic phenotype, cells were treated with the small molecule AdipoRon, a synthetic analogue of adiponectin. Although the responses varied between cells lines, they suggest a general influence of AdipoRon on metabolism, transport, immune system, cell stress and signalling. Summary: A stem cell based in vitro model of NAFLD recapitulates regulatory networks and suggests a steatosis associated phenotype. AdipoRon treatment influences metabolism, immune system, cell stress and signalling.
Collapse
Affiliation(s)
- Nina Graffmann
- Institute for Stem Cell Research and Regenerative Medicine, Heinrich Heine University Düsseldorf, Medical faculty, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Audrey Ncube
- Institute for Stem Cell Research and Regenerative Medicine, Heinrich Heine University Düsseldorf, Medical faculty, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Soraia Martins
- Institute for Stem Cell Research and Regenerative Medicine, Heinrich Heine University Düsseldorf, Medical faculty, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Aurelian Robert Fiszl
- Institute for Stem Cell Research and Regenerative Medicine, Heinrich Heine University Düsseldorf, Medical faculty, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Philipp Reuther
- Institute of Organic Chemistry and Macromolecular Chemistry, Heinrich-Heine University Düsseldorf 40225, Düsseldorf, Germany
| | - Martina Bohndorf
- Institute for Stem Cell Research and Regenerative Medicine, Heinrich Heine University Düsseldorf, Medical faculty, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Wasco Wruck
- Institute for Stem Cell Research and Regenerative Medicine, Heinrich Heine University Düsseldorf, Medical faculty, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Mathias Beller
- Institute for Mathematical Modeling of Biological Systems, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany.,Systems Biology of Lipid Metabolism, Heinrich-Heine University Düsseldorf 40225, Düsseldorf, Germany
| | - Constantin Czekelius
- Institute of Organic Chemistry and Macromolecular Chemistry, Heinrich-Heine University Düsseldorf 40225, Düsseldorf, Germany
| | - James Adjaye
- Institute for Stem Cell Research and Regenerative Medicine, Heinrich Heine University Düsseldorf, Medical faculty, Moorenstrasse 5, 40225 Düsseldorf, Germany
| |
Collapse
|
27
|
Mashek DG. Hepatic lipid droplets: A balancing act between energy storage and metabolic dysfunction in NAFLD. Mol Metab 2020; 50:101115. [PMID: 33186758 PMCID: PMC8324678 DOI: 10.1016/j.molmet.2020.101115] [Citation(s) in RCA: 126] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 10/21/2020] [Accepted: 11/06/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) is defined by the abundance of lipid droplets (LDs) in hepatocytes. While historically considered simply depots for energy storage, LDs are increasingly recognized to impact a wide range of biological processes that influence cellular metabolism, signaling, and function. While progress has been made toward understanding the factors leading to LD accumulation (i.e. steatosis) and its progression to advanced stages of NAFLD and/or systemic metabolic dysfunction, much remains to be resolved. SCOPE OF REVIEW This review covers many facets of LD biology. We provide a brief overview of the major pathways of lipid accretion and degradation that contribute to steatosis and how they are altered in NAFLD. The major focus is on the relationship between LDs and cell function and the detailed mechanisms that couple or uncouple steatosis from the severity and progression of NAFLD and systemic comorbidities. The importance of specific lipids and proteins within or on LDs as key components that determine whether LD accumulation is linked to cellular and metabolic dysfunction is presented. We discuss emerging areas of LD biology and future research directions that are needed to advance our understanding of the role of LDs in NAFLD etiology. MAJOR CONCLUSIONS Impairments in LD breakdown appear to contribute to disease progression, but inefficient incorporation of fatty acids (FAs) into LD-containing triacylglycerol (TAG) and the consequential changes in FA partitioning also affect NAFLD etiology. Increased LD abundance in hepatocytes does not necessarily equate to cellular dysfunction. While LD accumulation is the prerequisite step for most NAFLD cases, the protein and lipid composition of LDs are critical factors in determining the progression from simple steatosis. Further defining the detailed molecular mechanisms linking LDs to metabolic dysfunction is important for designing effective therapeutic approaches targeting NAFLD and its comorbidities.
Collapse
Affiliation(s)
- Douglas G Mashek
- Department of Biochemistry, Molecular Biology, and Biophysics, Department of Medicine, Division of Diabetes, Endocrinology, and Metabolism, University of Minnesota, Suite 6-155, 321 Church St. SE, Minneapolis, MN, 55455, USA.
| |
Collapse
|
28
|
Friend or Foe: Lipid Droplets as Organelles for Protein and Lipid Storage in Cellular Stress Response, Aging and Disease. Molecules 2020; 25:molecules25215053. [PMID: 33143278 PMCID: PMC7663626 DOI: 10.3390/molecules25215053] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/20/2020] [Accepted: 10/22/2020] [Indexed: 02/06/2023] Open
Abstract
Lipid droplets (LDs) were considered as a mere lipid storage organelle for a long time. Recent evidence suggests that LDs are in fact distinct and dynamic organelles with a specialized proteome and functions in many cellular roles. As such, LDs contribute to cellular signaling, protein and lipid homeostasis, metabolic diseases and inflammation. In line with the multitude of functions, LDs interact with many cellular organelles including mitochondria, peroxisomes, lysosomes, the endoplasmic reticulum and the nucleus. LDs are highly mobile and dynamic organelles and impaired motility disrupts the interaction with other organelles. The reduction of interorganelle contacts results in a multitude of pathophysiologies and frequently in neurodegenerative diseases. Contacts not only supply lipids for β-oxidation in mitochondria and peroxisomes, but also may include the transfer of toxic lipids as well as misfolded and harmful proteins to LDs. Furthermore, LDs assist in the removal of protein aggregates when severe proteotoxic stress overwhelms the proteasomal system. During imbalance of cellular lipid homeostasis, LDs also support cellular detoxification. Fine-tuning of LD function is of crucial importance and many diseases are associated with dysfunctional LDs. We summarize the current understanding of LDs and their interactions with organelles, providing a storage site for harmful proteins and lipids during cellular stress, aging inflammation and various disease states.
Collapse
|
29
|
Gunn PJ, Pramfalk C, Millar V, Cornfield T, Hutchinson M, Johnson EM, Nagarajan SR, Troncoso‐Rey P, Mithen RF, Pinnick KE, Traka MH, Green CJ, Hodson L. Modifying nutritional substrates induces macrovesicular lipid droplet accumulation and metabolic alterations in a cellular model of hepatic steatosis. Physiol Rep 2020; 8:e14482. [PMID: 32643289 PMCID: PMC7343665 DOI: 10.14814/phy2.14482] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 05/02/2020] [Accepted: 05/18/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND AND AIMS Nonalcoholic fatty liver disease (NAFLD) begins with steatosis, where a mixed macrovesicular pattern of large and small lipid droplets (LDs) develops. Since in vitro models recapitulating this are limited, the aims of this study were to develop mixed macrovesicular steatosis in immortalized hepatocytes and investigate effects on intracellular metabolism by altering nutritional substrates. METHODS Huh7 cells were cultured in 11 mM glucose and 2% human serum (HS) for 7 days before additional sugars and fatty acids (FAs), either with 200 µM FAs (low fat low sugar; LFLS), 5.5 mM fructose + 200 µM FAs (low fat high sugar; LFHS), or 5.5 mM fructose + 800 µM FAs (high fat high sugar; HFHS), were added for 7 days. FA metabolism, lipid droplet characteristics, and transcriptomic signatures were investigated. RESULTS Between the LFLS and LFHS conditions, there were few notable differences. In the HFHS condition, intracellular triacylglycerol (TAG) was increased and the LD pattern and distribution was similar to that found in primary steatotic hepatocytes. HFHS-treated cells had lower levels of de novo-derived FAs and secreted larger, TAG-rich lipoprotein particles. RNA sequencing and gene set enrichment analysis showed changes in several pathways including those involved in metabolism and cell cycle. CONCLUSIONS Repeated doses of HFHS treatment resulted in a cellular model of NAFLD with a mixed macrovesicular LD pattern and metabolic dysfunction. Since these nutrients have been implicated in the development of NAFLD in humans, the model provides a good physiological basis for studying NAFLD development or regression in vitro.
Collapse
Affiliation(s)
- Pippa J. Gunn
- Oxford Centre for Diabetes, Endocrinology and MetabolismRadcliffe Department of MedicineUniversity of OxfordOxfordUK
| | - Camilla Pramfalk
- Division of Clinical ChemistryDepartment of Laboratory MedicineKarolinska Institutet at Karolinska University Hospital HuddingeStockholmSweden
| | - Val Millar
- Target Discovery InstituteNuffield Department of MedicineUniversity of OxfordOxfordUK
| | - Thomas Cornfield
- Oxford Centre for Diabetes, Endocrinology and MetabolismRadcliffe Department of MedicineUniversity of OxfordOxfordUK
| | - Matthew Hutchinson
- Oxford Centre for Diabetes, Endocrinology and MetabolismRadcliffe Department of MedicineUniversity of OxfordOxfordUK
| | - Elspeth M. Johnson
- Oxford Centre for Diabetes, Endocrinology and MetabolismRadcliffe Department of MedicineUniversity of OxfordOxfordUK
| | - Shilpa R. Nagarajan
- Oxford Centre for Diabetes, Endocrinology and MetabolismRadcliffe Department of MedicineUniversity of OxfordOxfordUK
| | | | | | - Katherine E. Pinnick
- Oxford Centre for Diabetes, Endocrinology and MetabolismRadcliffe Department of MedicineUniversity of OxfordOxfordUK
| | | | - Charlotte J. Green
- Oxford Centre for Diabetes, Endocrinology and MetabolismRadcliffe Department of MedicineUniversity of OxfordOxfordUK
| | - Leanne Hodson
- Oxford Centre for Diabetes, Endocrinology and MetabolismRadcliffe Department of MedicineUniversity of OxfordOxfordUK
- National Institute for Health Research Oxford Biomedical Research CentreOxford University Hospital TrustsOxfordUK
| |
Collapse
|
30
|
Heinecke F, Mazzucco MB, Fornes D, Roberti S, Jawerbaum A, White V. The offspring from rats fed a fatty diet display impairments in the activation of liver peroxisome proliferator activated receptor alpha and features of fatty liver disease. Mol Cell Endocrinol 2020; 511:110818. [PMID: 32298755 DOI: 10.1016/j.mce.2020.110818] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 04/06/2020] [Accepted: 04/07/2020] [Indexed: 02/06/2023]
Abstract
Maternal obesity programs liver derangements similar to those of NAFLD. Our main goal was to evaluate whether these liver anomalies were related to aberrant PPARα function. Obesity was induced in female Albino-Wistar rats by a fatty diet (FD rats). Several parameters related to NAFLD were evaluated in both plasma and livers from fetuses of 21 days of gestation and 140-day-old offspring. FD fetuses and offspring developed increased levels of AST and ALT, signs of inflammation and oxidative and nitrative stress-related damage. FD offspring showed dysregulation of Plin2, CD36, Cyp4A, Aco, Cpt-1, Hadha and Acaa2 mRNA levels, genes involved in lipid metabolism and no catabolic effect of the PPARα agonist clofibrate. These results suggest that the FD offspring is prone to develop fatty liver, a susceptibility that can be linked to PPARα dysfunction, and that this could in turn be related to the liver impairments programmed by maternal obesity.
Collapse
Affiliation(s)
- Florencia Heinecke
- Laboratory of Reproduction and Metabolism, Centre for Pharmacological and Botanical Studies (CEFYBO-CONICET), School of Medicine University of Buenos Aires, Argentina
| | - María Belén Mazzucco
- Laboratory of Reproduction and Metabolism, Centre for Pharmacological and Botanical Studies (CEFYBO-CONICET), School of Medicine University of Buenos Aires, Argentina
| | - Daiana Fornes
- Laboratory of Reproduction and Metabolism, Centre for Pharmacological and Botanical Studies (CEFYBO-CONICET), School of Medicine University of Buenos Aires, Argentina
| | - Sabrina Roberti
- Laboratory of Reproduction and Metabolism, Centre for Pharmacological and Botanical Studies (CEFYBO-CONICET), School of Medicine University of Buenos Aires, Argentina
| | - Alicia Jawerbaum
- Laboratory of Reproduction and Metabolism, Centre for Pharmacological and Botanical Studies (CEFYBO-CONICET), School of Medicine University of Buenos Aires, Argentina
| | - Verónica White
- Laboratory of Reproduction and Metabolism, Centre for Pharmacological and Botanical Studies (CEFYBO-CONICET), School of Medicine University of Buenos Aires, Argentina.
| |
Collapse
|
31
|
Western diet induces severe nonalcoholic steatohepatitis, ductular reaction, and hepatic fibrosis in liver CGI-58 knockout mice. Sci Rep 2020; 10:4701. [PMID: 32170127 PMCID: PMC7070035 DOI: 10.1038/s41598-020-61473-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 02/27/2020] [Indexed: 12/13/2022] Open
Abstract
Humans and rodents with Comparative Gene Identification-58 (CGI-58) mutations manifest nonalcoholic fatty liver disease (NAFLD). Here we show that liver CGI-58 knockout (LivKO) mice fed a Western diet rapidly develop advanced NAFLD, including nonalcoholic steatohepatitis (NASH) and hepatic fibrosis. After 14 weeks of diet challenge, starting at 6 weeks of age, LivKO mice showed increased inflammatory cell infiltration and proinflammatory gene expression in the liver, which was associated with elevated plasma levels of aminotransferases. Hepatic ductular reactions, pericellular fibrosis, and bridging fibrosis were observed only in the LivKO mice. Consistently, the KO mice had a significant increase in hepatic mRNAs for fibrogenic genes. In addition, LivKO mice displayed massive accumulation of lipid droplets (LDs) in hepatocytes. LDs were also observed in the cholangiocytes of the LivKO mice, but not the floxed controls. Four of the five LD coat proteins, including perilipins 2, 3, 4, and 5, were increased in the CGI-58 KO liver. CRISPR/Cas9-mediated knockout of CGI-58 in Huh7 human hepatoma cells induced LD deposition and perilipin expression, suggesting a cell autonomous effect. Our findings establish the Western diet-fed LivKO mice as an animal model of NASH and hepatic fibrosis. These animals may facilitate preclinical screening of therapeutic agents that counter against NAFLD progression.
Collapse
|
32
|
Filipović N, Bočina I, Restović I, Grobe M, Kretzschmar G, Kević N, Mašek T, Vitlov Uljević M, Jurić M, Vukojević K, Saraga-Babić M, Vuica A. Ultrastructural characterization of vitamin D receptors and metabolizing enzymes in the lipid droplets of the fatty liver in rat. Acta Histochem 2020; 122:151502. [PMID: 31932064 DOI: 10.1016/j.acthis.2020.151502] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 11/11/2019] [Accepted: 11/11/2019] [Indexed: 01/10/2023]
Abstract
Vitamin D is a steroid hormone with numerous actions in the organism. There are strong evidences that relate vitamin D deficiency with liver lipid metabolism disturbances, but the mechanism of this action is still unknown. In our previous work we postulated the localization and accumulation of vitamin D receptor (VDR) in membrane of the lipid droplets (LDs) in hepatocytes. In this study, we applied the transmission electron microscopy (TEM) to confirm this hypothesis by using a long-term (6 months) high sucrose intake rat model that was previously found to be appropriate for research of the hepatic lipid accumulation. In addition to the VDR, we also found key vitamin D metabolizing enzymes, 1α-hydroxylase and CYP 24 associated with the membrane of the LDs. A light-microscopy data revealed significant increase in expression of VDR and CYP 24 in liver of high-sucrose treated rats, in comparison to controlones. According to the best of our knowledge, this is a first study confirming the presence of the VDR in the membrane of the LDs in general and also in particular in LDs of the hepatocytes that were accumulated as a consequence of the prolonged high sucrose intake. Moreover, we found association of main vitamin D metabolizing enzymes with LD membrane. These results provide a new insight in the possible relation of vitamin D signalling system with LD morphology and function and with the lipid metabolism in general.
Collapse
|
33
|
Zhou J, Cui S, He Q, Guo Y, Pan X, Zhang P, Huang N, Ge C, Wang G, Gonzalez FJ, Wang H, Hao H. SUMOylation inhibitors synergize with FXR agonists in combating liver fibrosis. Nat Commun 2020; 11:240. [PMID: 31932588 PMCID: PMC6957516 DOI: 10.1038/s41467-019-14138-6] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 12/19/2019] [Indexed: 02/06/2023] Open
Abstract
Farnesoid X receptor (FXR) is a promising target for nonalcoholic steatohepatitis (NASH) and fibrosis. Although various FXR agonists have shown anti-fibrotic effects in diverse preclinical animal models, the response rate and efficacies in clinical trials were not optimum. Here we report that prophylactic but not therapeutic administration of obeticholic acid (OCA) prevents hepatic stellate cell (HSC) activation and fibrogenesis. Activated HSCs show limited response to OCA and other FXR agonists due to enhanced FXR SUMOylation. SUMOylation inhibitors rescue FXR signaling and thereby increasing the efficacy of OCA against HSC activation and fibrosis. FXR upregulates Perilipin-1, a direct target gene of FXR, to stabilize lipid droplets and thereby prevent HSC activation. Therapeutic coadministration of OCA and SUMOylation inhibitors drastically impedes liver fibrosis induced by CCl4, bile duct ligation, and more importantly NASH. In conclusion, we propose a promising therapeutic approach by combining SUMOylation inhibitors and FXR agonists for liver fibrosis.
Collapse
Affiliation(s)
- Jiyu Zhou
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, 210009, Nanjing, China
| | - Shuang Cui
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, 210009, Nanjing, China
| | - Qingxian He
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, 210009, Nanjing, China
| | - Yitong Guo
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, 210009, Nanjing, China
| | - Xiaojie Pan
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, 210009, Nanjing, China
| | - Pengfei Zhang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, 210009, Nanjing, China
| | - Ningning Huang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, 210009, Nanjing, China
| | - Chaoliang Ge
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, 210009, Nanjing, China
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, 230022, Hefei, China
| | - Guangji Wang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, 210009, Nanjing, China.
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Hong Wang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, 210009, Nanjing, China.
| | - Haiping Hao
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, 210009, Nanjing, China.
| |
Collapse
|
34
|
Kubota N, Ojima H, Hatano M, Yamazaki K, Masugi Y, Tsujikawa H, Fujii-Nishimura Y, Ueno A, Kurebayashi Y, Shinoda M, Kitago M, Abe Y, Kitagawa Y, Sakamoto M. Clinicopathological features of hepatocellular carcinoma with fatty change: Tumors with macrovesicular steatosis have better prognosis and aberrant expression patterns of perilipin and adipophilin. Pathol Int 2020; 70:199-209. [PMID: 31930673 DOI: 10.1111/pin.12889] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 11/26/2019] [Indexed: 12/19/2022]
Abstract
The clinicopathological characteristics of steatosis in hepatocellular carcinoma (HCC) remain unclear. Here, we elucidate the features of macrovesicular steatosis (MaS) and microvesicular steatosis (MiS) in HCC and their relationships with background liver steatosis. A total of 165 HCC lesions were classified as MaS-HCC, MiS-HCC, or conventional HCC (cHCC) according to the cutoff value of 30% MaS or MiS in tumor cells. We analyzed the clinicopathological differences among these groups. MaS-HCC had less portal vein invasion, a higher proportion of HCC with intratumoral fibrosis, and a lower cumulative risk of recurrence than MiS-HCC or cHCC. Moreover, both MaS-HCC and MiS-HCC had lower incidences of hepatitis virus infection and higher levels of HbA1c than cHCC. Background liver steatosis was also higher in MaS-HCC than in cHCC. Immunohistochemical expression of perilipin (Plin1) and adipophilin (ADRP), major proteins expressed on lipid droplet membranes, revealed that almost all lipid droplets in HCC were Plin1 negative, whereas those in background liver were positive. In contrast, ADRP was expressed on lipid droplets in both HCC and background liver. We concluded that MaS-HCC and MiS-HCC were associated with metabolic abnormalities but exhibited different biologic behaviors. Furthermore, lipid droplets in HCC were pathophysiologically different from those in background liver.
Collapse
Affiliation(s)
- Naoto Kubota
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Hidenori Ojima
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Mami Hatano
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan.,Department of Pathology, National Hospital Organization, Tokyo Medical Center, Tokyo, Japan
| | - Ken Yamazaki
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Yohei Masugi
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Hanako Tsujikawa
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | | | - Akihisa Ueno
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Yutaka Kurebayashi
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Masahiro Shinoda
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Minoru Kitago
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Yuta Abe
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Yuko Kitagawa
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Michiie Sakamoto
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
35
|
Yu L, Li Y, Grisé A, Wang H. CGI-58: Versatile Regulator of Intracellular Lipid Droplet Homeostasis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1276:197-222. [PMID: 32705602 PMCID: PMC8063591 DOI: 10.1007/978-981-15-6082-8_13] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Comparative gene identification-58 (CGI-58), also known as α/β-hydrolase domain-containing 5 (ABHD5), is a member of a large family of proteins containing an α/β-hydrolase-fold. CGI-58 is well-known as the co-activator of adipose triglyceride lipase (ATGL), which is a key enzyme initiating cytosolic lipid droplet lipolysis. Mutations in either the human CGI-58 or ATGL gene cause an autosomal recessive neutral lipid storage disease, characterized by the excessive accumulation of triglyceride (TAG)-rich lipid droplets in the cytoplasm of almost all cell types. CGI-58, however, has ATGL-independent functions. Distinct phenotypes associated with CGI-58 deficiency commonly include ichthyosis (scaly dry skin), nonalcoholic steatohepatitis, and hepatic fibrosis. Through regulated interactions with multiple protein families, CGI-58 controls many metabolic and signaling pathways, such as lipid and glucose metabolism, energy balance, insulin signaling, inflammatory responses, and thermogenesis. Recent studies have shown that CGI-58 regulates the pathogenesis of common metabolic diseases in a tissue-specific manner. Future studies are needed to molecularly define ATGL-independent functions of CGI-58, including the newly identified serine protease activity of CGI-58. Elucidation of these versatile functions of CGI-58 may uncover fundamental cellular processes governing lipid and energy homeostasis, which may help develop novel approaches that counter against obesity and its associated metabolic sequelae.
Collapse
Affiliation(s)
- Liqing Yu
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Yi Li
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Alison Grisé
- College of Computer, Math, and Natural Sciences, College of Behavioral and Social Sciences, University of Maryland, College Park, MD, USA
| | - Huan Wang
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
36
|
Hodson L, Gunn PJ. The regulation of hepatic fatty acid synthesis and partitioning: the effect of nutritional state. Nat Rev Endocrinol 2019; 15:689-700. [PMID: 31554932 DOI: 10.1038/s41574-019-0256-9] [Citation(s) in RCA: 167] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is an increasing global public health burden. NAFLD is strongly associated with type 2 diabetes mellitus, obesity and cardiovascular disease and begins with intrahepatic triacylglycerol accumulation. Under healthy conditions, the liver regulates lipid metabolism to meet systemic energy needs in the fed and fasted states. The processes of fatty acid uptake, fatty acid synthesis and the intracellular partitioning of fatty acids into storage, oxidation and secretion pathways are tightly regulated. When one or more of these processes becomes dysregulated, excess lipid accumulation can occur. Although genetic and environmental factors have been implicated in the development of NAFLD, it remains unclear why an imbalance in these pathways begins. The regulation of fatty acid partitioning occurs at several points, including during triacylglycerol synthesis, lipid droplet formation and lipolysis. These processes are influenced by enzyme function, intake of dietary fats and sugars and whole-body metabolism, and are further affected by the presence of obesity or insulin resistance. Insight into how the liver controls fatty acid metabolism in health and how these processes might be affected in disease would offer the potential for new therapeutic treatments for NAFLD to be developed.
Collapse
Affiliation(s)
- Leanne Hodson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Headington, Oxford, UK.
- Oxford NIHR Biomedical Research Centre, Churchill Hospital, Headington, Oxford, UK.
| | - Pippa J Gunn
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Headington, Oxford, UK
- Oxford NIHR Biomedical Research Centre, Churchill Hospital, Headington, Oxford, UK
| |
Collapse
|
37
|
Hidalgo I, Nájera N, Meaney E, Pérez-Durán J, Valdespino-Vazquez Y, Villarreal F, Ceballos G. Effects of (-)-epicatechin on the time course of the expression of perilipins in a diet-induced model of nonalcoholic steatohepatitis. J Nutr Biochem 2019; 77:108296. [PMID: 32007822 DOI: 10.1016/j.jnutbio.2019.108296] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 10/30/2019] [Accepted: 11/07/2019] [Indexed: 12/25/2022]
Abstract
The existing treatments for nonalcoholic steatohepatitis (NASH) are not completely effective. The need for new alternatives without adverse effects and low cost, such as the flavonoid (-)-epicatechin (EC), which has beneficial effects on lipid metabolism and cardiovascular diseases, arises. The objective of this work was to analyze EC effects in the NASH induced by a Paigen-type diet (PD). Mice were administered with (1) normal chow and water, (2) PD + fructose 30% and (3) PD + fructose 30% + EC (1 mg/kg) per gavage during 9 weeks. At the end of each treatment, serum was collected for analysis of the biochemical profile and liver enzymes. The liver was collected for microscopic analysis and for the evaluation of the relative expression of Plin2, Plin3, CD36, adiponectin and UCP2. Results showed that EC reduced weight gain and decreased triglyceride (TG), low-density lipoprotein cholesterol, TG/high-density lipoprotein and the activity of liver enzymes (alanine aminotransferase and alkaline phosphatase), suggesting lower liver damage. The microscopic analysis showed less "balloonization" of the hepatocyte, small drops of lipids, less accumulation of collagen and infiltration of inflammatory cells as compared to nontreated group. Finally, a decrease in the expression of Plin 2 was observed. While CD36 decreased, adiponectin and UCP2 increased. In conclusion, EC improves the biochemical profile, the microscopic characteristics and protein expression. Therefore, it may be a possible therapeutic approach for NASH since it prevents the progression of the hepatic and metabolic damage induced by high-fat diets.
Collapse
Affiliation(s)
- Isabel Hidalgo
- Seccion de Posgrado, Escuela Superior de Medicina, Instituto Politecnico Nacional, Salvador Díaz Mirón esq. Plan de San Luis S/N, Miguel Hidalgo, Casco de Santo Tomas, 11340 Ciudad de Mexico, CDMX
| | - Nayelli Nájera
- Seccion de Posgrado, Escuela Superior de Medicina, Instituto Politecnico Nacional, Salvador Díaz Mirón esq. Plan de San Luis S/N, Miguel Hidalgo, Casco de Santo Tomas, 11340 Ciudad de Mexico, CDMX
| | - Eduardo Meaney
- Seccion de Posgrado, Escuela Superior de Medicina, Instituto Politecnico Nacional, Salvador Díaz Mirón esq. Plan de San Luis S/N, Miguel Hidalgo, Casco de Santo Tomas, 11340 Ciudad de Mexico, CDMX
| | - Javier Pérez-Durán
- Laboratorio de Genetica y Genomica humana, Instituto Nacional de Perinatologia, Montes Urales 800, Lomas Virreyes, Lomas de Chapultepec, 11000 Ciudad de Mexico, CDMX
| | - Yolotzin Valdespino-Vazquez
- Anatomia Patologica. Instituto Nacional de Perinatologia, Montes Urales 800, Lomas Virreyes, Lomas de Chapultepec, 11000 Ciudad de Mexico, CDMX
| | - Francisco Villarreal
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA
| | - Guillermo Ceballos
- Seccion de Posgrado, Escuela Superior de Medicina, Instituto Politecnico Nacional, Salvador Díaz Mirón esq. Plan de San Luis S/N, Miguel Hidalgo, Casco de Santo Tomas, 11340 Ciudad de Mexico, CDMX.
| |
Collapse
|
38
|
Straub BK, Witzel HR, Pawella LM, Renner M, Eiteneuer E, Hashani M, Schirmacher P, Roth W, Mechtersheimer G. Perilipin 1 Expression Differentiates Liposarcoma from Other Types of Soft Tissue Sarcoma. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:1547-1558. [DOI: 10.1016/j.ajpath.2019.04.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 03/19/2019] [Accepted: 04/10/2019] [Indexed: 11/28/2022]
|
39
|
Tian S, Lei P, Teng C, Sun Y, Song X, Li B, Shan Y. Targeting PLIN2/PLIN5-PPARγ: Sulforaphane Disturbs the Maturation of Lipid Droplets. Mol Nutr Food Res 2019; 63:e1900183. [PMID: 31325205 DOI: 10.1002/mnfr.201900183] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 07/08/2019] [Indexed: 01/05/2023]
Abstract
SCOPE The effects of sulforaphane (SFN) on the maturation of lipid droplets (LDs)-the storage units for free fatty acids and sterols as triacylglycerides (TAG) and cholesterol esters (CE)-are far from being understood, despite the fact that SFN is known to be beneficial for ameliorating lipid metabolism disorders. METHODS AND RESULTS High-fat-intake models are established in both HHL-5 hepatocytes and rodents. The numbers and sizes of LDs are decreased by SFN. The accumulation of lipid core components (TAG & CE) is reduced and the expression of their key synthetases, acyl-coenzyme A: diacylglycerol acyltransferases 2 (DGAT2) and acyl-coenzyme A: cholesterol acyltransferases 1 (ACAT1), is also inhibited. Moreover, SFN decreases LD-associated protein PLIN2 and PLIN5 expression, but not that of PLIN1 and PLIN3, both in vivo and in vitro. Furthermore, over-expression of peroxisome proliferator-activated receptor gamma (PPARγ) induces the accumulation of TAG and the up-regulation of PLIN2 and PLIN5, which are not reversed by SFN. These results suggest that PPARγ may be a target of SFN in lipid metabolism. CONCLUSION SFN disturbs LD maturation by inhibiting the formation of the neutral lipid core and decreases PLIN2 and PLIN5 via down-regulation of PPARγ.
Collapse
Affiliation(s)
- Sicong Tian
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, 325035, China
| | - Peng Lei
- Department of Food Science and Engineering, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China
| | - Chunying Teng
- Department of Food Science and Engineering, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China
| | - Yao Sun
- Department of Food Science and Engineering, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China
| | - Xinyue Song
- Department of Food Science and Engineering, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China
| | - Baolong Li
- Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Yujuan Shan
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, 325035, China
| |
Collapse
|
40
|
Gu Y, Yang Y, Cao X, Zhao Y, Gao X, Sun C, Zhang F, Yuan Y, Xu Y, Zhang J, Xiao L, Ye J. Plin3 protects against alcoholic liver injury by facilitating lipid export from the endoplasmic reticulum. J Cell Biochem 2019; 120:16075-16087. [PMID: 31119787 DOI: 10.1002/jcb.28889] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 03/01/2019] [Accepted: 03/15/2019] [Indexed: 12/20/2022]
Abstract
Hepatic lipid accumulation is the most common pathological characteristic of alcoholic liver disease (ALD). In mammalian cells, excess neutral lipids are stored in lipid droplets (LDs). As a member of perilipin family proteins, Plin3 was recently found to regulate the LD biogenesis. However, the roles and mechanism of Plin3 in ALD progression remain unclear. Herein, we found that alcohol stimulated Plin3 expression in both mouse livers and cultured AML12 mouse hepatic cells, which was accompanied by excess LD accumulation in hepatocytes. The elevations of Plin3 in alcohol-treated hepatocytes paralleled with the levels of both PPARα and γ, and the protein degradation of Plin3 was also reduced after alcohol exposure. Moreover, Plin3 knockdown increased cellular sensitivity to alcohol-induced apoptosis, endoplasmic reticulum (ER) stress, and inflammatory cytokines release, including TNF-α, IL-1, and IL-6β. Notably, alcohol exacerbated triglycerides (TG) accumulation in the ER and caused ER dilation in Plin3-knockdown AML12 cells. Finally, we observed that Plin3 interacted with dynein subunit Dync1i1 and mediated the colocalization of LDs and microtubules, while high concentration of alcohol disrupted microtubules and caused dispersion of excess small LDs in cytoplasm. Summarily, Plin3 promotes lipid export from the ER and reduces ER lipotoxic stress, thereby, protecting against alcoholic liver injury. Moreover, Plin3 could be an adapter protein mediating LD transport by microtubules. This study explored the roles of Plin3 in alcohol-induced hepatic injury, suggesting Plin3 as a potential target for the prevention of ALD progression.
Collapse
Affiliation(s)
- Yu Gu
- State Key Laboratory of Cancer Biology and Department of Pathology, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China.,Department of Pathology, Basic Medicine School, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Ying Yang
- Department of Pathology, Basic Medicine School, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Xiangmei Cao
- Department of Pathology, Basic Medical School, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Yuanlin Zhao
- State Key Laboratory of Cancer Biology and Department of Pathology, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Xing Gao
- State Key Laboratory of Cancer Biology and Department of Pathology, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Chao Sun
- State Key Laboratory of Cancer Biology and Department of Pathology, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China.,Department of Neurology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Feng Zhang
- State Key Laboratory of Cancer Biology and Department of Pathology, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China.,Department of Pathology, Basic Medicine School, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yuan Yuan
- Department of Pathology, Basic Medicine School, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yuqiao Xu
- State Key Laboratory of Cancer Biology and Department of Pathology, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China.,Department of Pathology, Basic Medicine School, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jin Zhang
- Department of Pathology, Basic Medicine School, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Liming Xiao
- State Key Laboratory of Cancer Biology and Department of Pathology, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jing Ye
- State Key Laboratory of Cancer Biology and Department of Pathology, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China.,Department of Pathology, Basic Medicine School, The Fourth Military Medical University, Xi'an, Shaanxi, China
| |
Collapse
|
41
|
Zeng H, Guo X, Zhou F, Xiao L, Liu J, Jiang C, Xing M, Yao P. Quercetin alleviates ethanol-induced liver steatosis associated with improvement of lipophagy. Food Chem Toxicol 2018; 125:21-28. [PMID: 30580029 DOI: 10.1016/j.fct.2018.12.028] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 12/06/2018] [Accepted: 12/18/2018] [Indexed: 02/07/2023]
Abstract
Although emerging evidence demonstrated that quercetin could be explored as a potential candidate for the early intervention of alcoholic liver disease (ALD), the exact mechanisms against ethanol-induced hepatic steatosis haven't been fully elucidated. Herein, we investigated the effect of quercetin on liver steatosis caused by chronic-plus-single-binge ethanol feeding, focusing on lipophagy. Adult male mice were pair-fed with liquid diets containing ethanol (28% of total calories) and treated with quercetin for 12 weeks. Chronic-plus-binge ethanol consumption led to lipid droplets accumulation and liver damage as evidenced by histopathological changes, the increased content of triglyceride in serum and liver, and the elevated of serum ALT and AST level, which were greatly attenuated by quercetin. Moreover, quercetin blocked autophagy suppression by chronic-binge ethanol intake as manifested by the morphological improvement of mitochondrial characteristics, the increased number of autolysosome and restoration of autophagy-related protein expression. Furthermore, quercetin promoted lipophagy confirmed by the decreased perilipin 2 (PLIN2) level, activated AMPK activity and increased co-localization of liver LC3II and PLIN2 proteins. Collectively, these findings suggest that regular consumption of dietary quercetin has a role in preventing hepatic steatosis induced by chronic-plus-binge ethanol feeding, which mechanism may associate with the evident regulatory effect of quercetin on lipophagy.
Collapse
Affiliation(s)
- Hongmei Zeng
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; Ministry of Education Key Laboratory of Environment, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiaoping Guo
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; Ministry of Education Key Laboratory of Environment, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Feng Zhou
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; Ministry of Education Key Laboratory of Environment, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Lin Xiao
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; Ministry of Education Key Laboratory of Environment, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jingjing Liu
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; Ministry of Education Key Laboratory of Environment, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Chunjie Jiang
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; Ministry of Education Key Laboratory of Environment, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Mingyou Xing
- Department of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Ping Yao
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; Ministry of Education Key Laboratory of Environment, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
42
|
Arisqueta L, Navarro-Imaz H, Labiano I, Rueda Y, Fresnedo O. High-fat diet overfeeding promotes nondetrimental liver steatosis in female mice. Am J Physiol Gastrointest Liver Physiol 2018; 315:G772-G780. [PMID: 30095299 DOI: 10.1152/ajpgi.00022.2018] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
High-fat diet (HFD) feeding or leptin-deficient mice are extensively used as models resembling features of human nonalcoholic fatty liver disease (NAFLD). The concurrence of experimental factors as fat content and source or total caloric intake leads to prominent differences in the development of the hepatic steatosis and related disturbances. In this work, we characterized the hepatic lipid accumulation induced by HFD in wild-type (WT) and ob/ ob mice with the purpose of differentiating adaptations to HFD from those specific of increased overfeeding due to leptin deficiency-associated hyperphagia. Given that most published works have been done in male models, we used female mice with the aim of increasing the body of evidence regarding NAFLD in female subjects. HFD promoted liver lipid accumulation only in the hyperphagic strain. Nevertheless, a decrease of lipid droplet-associated cholesteryl ester (CE) in both WT and obese animals was observed. These changes were accompanied by an improvement in the profile of lipoproteins that transport cholesterol and liver function markers in plasma from ob/ ob mice and a lower hepatic index. Using primary hepatocytes from female mice, overaccumulation of CE induced by 0.4 mM oleic acid reversed in the presence of a specific Takeda G protein-coupled bile acid receptor agonist. Nevertheless, hepatocytes from male mice were not responsive. This study suggests that enterohepatic circulation of bile acids might be one of the factors that can affect sex dimorphism in NAFLD development, which underlines the importance of including female models in the NAFLD research field. NEW & NOTEWORTHY This work provides new insight into the use of high-fat diet as a model to induce nonalcoholic fatty liver disease in wild-type and ob/ ob female mice. We show that high-fat diet induces steatosis only in ob/ ob mice while, surprisingly, several health indicators improve. Noteworthy, experiments with primary hepatocytes from male and female mice show that they express Takeda G protein-coupled bile acid receptor and that it and bile acid enterohepatic circulation might be accountable for sex dimorphism in nonalcoholic fatty liver disease development.
Collapse
Affiliation(s)
- Lino Arisqueta
- Facultad de Ciencias Naturales y Ambientales, Universidad Internacional SEK , Quito , Ecuador
| | - Hiart Navarro-Imaz
- Lipids and Liver Research Group, Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, Leioa, Spain
| | - Ibone Labiano
- Department of Liver and Gastrointestinal Diseases, Health Research Institute, Biodonostia, Spain
| | - Yuri Rueda
- Lipids and Liver Research Group, Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, Leioa, Spain
| | - Olatz Fresnedo
- Lipids and Liver Research Group, Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, Leioa, Spain
| |
Collapse
|
43
|
Hoang NA, Richter F, Schubert M, Lorkowski S, Klotz LO, Steinbrenner H. Differential capability of metabolic substrates to promote hepatocellular lipid accumulation. Eur J Nutr 2018; 58:3023-3034. [PMID: 30368556 DOI: 10.1007/s00394-018-1847-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 10/23/2018] [Indexed: 02/07/2023]
Abstract
PURPOSE Excessive storage of triacylglycerides (TAGs) in lipid droplets within hepatocytes is a hallmark of non-alcoholic fatty liver disease (NAFLD), one of the most widespread metabolic disorders in Western societies. For the purpose of exploring molecular pathways in NAFLD development and testing potential drug candidates, well-characterised experimental models of ectopic TAG storage in hepatocytes are needed. METHODS Using an optimised Oil Red O assay, immunoblotting and real-time qRT-PCR, we compared the capability of dietary monosaccharides and fatty acids to promote lipid accumulation in HepG2 human hepatoma cells. RESULTS Both high glucose and high fructose resulted in intracellular lipid accumulation after 48 h, and this was further augmented (up to twofold, as compared to basal levels) by co-treatment with the lipogenesis-stimulating hormone insulin and the pro-inflammatory cytokine tumour necrosis factor alpha (TNF-α), respectively. The fatty acids palmitic and oleic acid were even more effective than these carbohydrates, inducing significantly elevated TAG storage already after 24 h of treatment. Highest (about threefold) increases in lipid accumulation were observed upon treatment with oleic acid, alone as well as in combinations with palmitic acid or with high glucose and insulin. Increases in protein levels of a major lipid droplet coat protein, perilipin-2 (PLIN2), mirrored intracellular lipid accumulation following different treatment regimens. CONCLUSIONS Several treatment regimens of excessive fat and sugar supply promoted lipid accumulation in HepG2 cells, albeit with differences in the extent and rapidity of steatogenesis. PLIN2 is a candidate molecular marker of sustained lipid accumulation in HepG2 cells.
Collapse
Affiliation(s)
- Ngoc Anh Hoang
- Institute of Nutritional Sciences, Nutrigenomics, Friedrich-Schiller-Universität Jena, Jena, Germany
| | - Friederike Richter
- Institute of Nutritional Sciences, Nutrigenomics, Friedrich-Schiller-Universität Jena, Jena, Germany
| | - Martin Schubert
- Department of Nutritional Biochemistry and Physiology, Institute of Nutritional Sciences, Friedrich-Schiller-Universität Jena, Jena, Germany
- Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD) Halle-Jena-Leipzig, Jena, Germany
| | - Stefan Lorkowski
- Department of Nutritional Biochemistry and Physiology, Institute of Nutritional Sciences, Friedrich-Schiller-Universität Jena, Jena, Germany
- Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD) Halle-Jena-Leipzig, Jena, Germany
| | - Lars-Oliver Klotz
- Institute of Nutritional Sciences, Nutrigenomics, Friedrich-Schiller-Universität Jena, Jena, Germany
| | - Holger Steinbrenner
- Institute of Nutritional Sciences, Nutrigenomics, Friedrich-Schiller-Universität Jena, Jena, Germany.
| |
Collapse
|
44
|
Lenz D, McClean P, Kansu A, Bonnen PE, Ranucci G, Thiel C, Straub BK, Harting I, Alhaddad B, Dimitrov B, Kotzaeridou U, Wenning D, Iorio R, Himes RW, Kuloğlu Z, Blakely EL, Taylor RW, Meitinger T, Kölker S, Prokisch H, Hoffmann GF, Haack TB, Staufner C. SCYL1 variants cause a syndrome with low γ-glutamyl-transferase cholestasis, acute liver failure, and neurodegeneration (CALFAN). Genet Med 2018; 20:1255-1265. [PMID: 29419818 PMCID: PMC5989927 DOI: 10.1038/gim.2017.260] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 11/27/2017] [Indexed: 02/07/2023] Open
Abstract
PURPOSE Biallelic mutations in SCYL1 were recently identified as causing a syndromal disorder characterized by peripheral neuropathy, cerebellar atrophy, ataxia, and recurrent episodes of liver failure. The occurrence of SCYL1 deficiency among patients with previously undetermined infantile cholestasis or acute liver failure has not been studied; furthermore, little is known regarding the hepatic phenotype. METHODS We aimed to identify patients with SCYL1 variants within an exome-sequencing study of individuals with infantile cholestasis or acute liver failure of unknown etiology. Deep clinical and biochemical phenotyping plus analysis of liver biopsies and functional studies on fibroblasts were performed. RESULTS Seven patients from five families with biallelic SCYL1 variants were identified. The main clinical phenotype was recurrent low γ-glutamyl-transferase (GGT) cholestasis or acute liver failure with onset in infancy and a variable neurological phenotype of later onset (CALFAN syndrome). Liver crises were triggered by febrile infections and were transient, but fibrosis developed. Functional studies emphasize that SCYL1 deficiency is linked to impaired intracellular trafficking. CONCLUSION SCYL1 deficiency can cause recurrent low-GGT cholestatic liver dysfunction in conjunction with a variable neurological phenotype. Like NBAS deficiency, it is a member of the emerging group of congenital disorders of intracellular trafficking causing hepatopathy.
Collapse
Affiliation(s)
- Dominic Lenz
- Division of Neuropediatrics and Pediatric Metabolic Medicine, Department of General Pediatrics, University Hospital Heidelberg, Heidelberg, Germany
| | | | - Aydan Kansu
- Division of Pediatric Gastroenterology, Department of Pediatrics, Ankara University School of Medicine, Ankara, Turkey
| | - Penelope E Bonnen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Giusy Ranucci
- Department of Translational Medical Sciences, Section of Pediatrics, Liver Unit, University of Naples Federico II, Italy, Italy
- Division of Metabolism, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Christian Thiel
- Division of Neuropediatrics and Pediatric Metabolic Medicine, Department of General Pediatrics, University Hospital Heidelberg, Heidelberg, Germany
| | - Beate K Straub
- Institutes of Pathology, University Hospital Heidelberg, Heidelberg, Germany
- Institute of Pathology and Tissue Bank of the, University Medical Center Mainz, Mainz, Germany
| | - Inga Harting
- Department of Neuroradiology, University Hospital Heidelberg, Heidelberg, Germany
| | - Bader Alhaddad
- Institute of Human Genetics, Technische Universität München, Munich, Germany
| | - Bianca Dimitrov
- Division of Neuropediatrics and Pediatric Metabolic Medicine, Department of General Pediatrics, University Hospital Heidelberg, Heidelberg, Germany
| | - Urania Kotzaeridou
- Division of Neuropediatrics and Pediatric Metabolic Medicine, Department of General Pediatrics, University Hospital Heidelberg, Heidelberg, Germany
| | - Daniel Wenning
- Division of Neuropediatrics and Pediatric Metabolic Medicine, Department of General Pediatrics, University Hospital Heidelberg, Heidelberg, Germany
| | - Raffaele Iorio
- Department of Translational Medical Sciences, Section of Pediatrics, Liver Unit, University of Naples Federico II, Italy, Italy
| | - Ryan W Himes
- Department of Pediatrics, Section of Gastroenterology, Hepatology, and Nutrition, Texas Children's Hospital, Houston, Texas, USA
| | - Zarife Kuloğlu
- Division of Pediatric Gastroenterology, Department of Pediatrics, Ankara University School of Medicine, Ankara, Turkey
| | - Emma L Blakely
- Wellcome Centre for Mitochondrial Research, Institute of Neuroscience, The Medical School, Newcastle University, Newcastle upon Tyne, UK
| | - Robert W Taylor
- Wellcome Centre for Mitochondrial Research, Institute of Neuroscience, The Medical School, Newcastle University, Newcastle upon Tyne, UK
| | - Thomas Meitinger
- Institute of Human Genetics, Technische Universität München, Munich, Germany
- Institute of Human Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Stefan Kölker
- Division of Neuropediatrics and Pediatric Metabolic Medicine, Department of General Pediatrics, University Hospital Heidelberg, Heidelberg, Germany
| | - Holger Prokisch
- Institute of Human Genetics, Technische Universität München, Munich, Germany
- Institute of Human Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Georg F Hoffmann
- Division of Neuropediatrics and Pediatric Metabolic Medicine, Department of General Pediatrics, University Hospital Heidelberg, Heidelberg, Germany
| | - Tobias B Haack
- Institute of Human Genetics, Technische Universität München, Munich, Germany
- Institute of Human Genetics, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Christian Staufner
- Division of Neuropediatrics and Pediatric Metabolic Medicine, Department of General Pediatrics, University Hospital Heidelberg, Heidelberg, Germany.
| |
Collapse
|
45
|
Possible Involvement of Mitochondrial Dysfunction and Oxidative Stress in a Cellular Model of NAFLD Progression Induced by Benzo[a]pyrene/Ethanol CoExposure. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:4396403. [PMID: 30147834 PMCID: PMC6083493 DOI: 10.1155/2018/4396403] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 06/18/2018] [Accepted: 06/26/2018] [Indexed: 12/11/2022]
Abstract
Exposure to xenobiotics could favor the transition of nonalcoholic fatty liver (NAFL) to nonalcoholic steatohepatitis in obese patients. Recently, we showed in different models of NAFL that benzo[a]pyrene (B[a]P) and ethanol coexposure induced a steatohepatitis-like state. One model was HepaRG cells incubated with stearate and oleate for 2 weeks. In the present study, we wished to determine in this model whether mitochondrial dysfunction and reactive oxygen species (ROS) overproduction could be involved in the occurrence of this steatohepatitis-like state. CRISPR/Cas9-modified cells were also used to specify the role of aryl hydrocarbon receptor (AhR), which is potently activated by B[a]P. Thus, nonsteatotic and steatotic HepaRG cells were treated with B[a]P, ethanol, or both molecules for 2 weeks. B[a]P/ethanol coexposure reduced mitochondrial respiratory chain activity, mitochondrial respiration, and mitochondrial DNA levels and induced ROS overproduction in steatotic HepaRG cells. These deleterious effects were less marked or absent in steatotic cells treated with B[a]P alone or ethanol alone and in nonsteatotic cells treated with B[a]P/ethanol. Our study also disclosed that B[a]P/ethanol-induced impairment of mitochondrial respiration was dependent on AhR activation. Hence, mitochondrial dysfunction and ROS generation could explain the occurrence of a steatohepatitis-like state in steatotic HepaRG cells exposed to B[a]P and ethanol.
Collapse
|
46
|
Hashani M, Witzel HR, Pawella LM, Lehmann-Koch J, Schumacher J, Mechtersheimer G, Schnölzer M, Schirmacher P, Roth W, Straub BK. Widespread expression of perilipin 5 in normal human tissues and in diseases is restricted to distinct lipid droplet subpopulations. Cell Tissue Res 2018; 374:121-136. [PMID: 29752569 DOI: 10.1007/s00441-018-2845-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 04/18/2018] [Indexed: 12/18/2022]
Abstract
Diseases associated with the accumulation of lipid droplets are increasing in western countries. Lipid droplet biogenesis, structure and degradation are regulated by proteins of the perilipin family. Perilipin 5 has been shown to regulate basal lipolysis in oxidative tissues. We examine perilipin 5 in normal human tissues and in diseases using protein biochemical and microscopic techniques. Perilipin 5 was constitutively located at small lipid droplets in skeletal myocytes, cardiomyocytes and brown adipocytes. In addition, perilipin 5 was detected in the epithelia of the gastrointestinal and urogenital tract, especially in hepatocytes, the mitochondria-rich parietal cells of the stomach, tubular kidney cells and ductal cells of the salivary gland and pancreas. Granular cytoplasmic expression, without a lipid droplet-bound localization was detected elsewhere. In cardiomyopathies, in skeletal muscle diseases and during hepatocyte steatogenesis, perilipin 5 was upregulated and localized to larger and more numerous lipid droplets. In steatotic human hepatocytes, perilipin 5 was moderately increased and colocalized with perilipins 1 and 2 but not with perilipin 3 at lipid droplets. In liver diseases implicated in alterations of mitochondria, such as mitochondriopathies, alcoholic liver disease, Wilson's disease and acute liver injury, perilipin 5 was frequently localized to small lipid droplets and less in the cytoplasm. In tumorigenesis, perilipin 5 was especially upregulated in lipo-, leio- and rhabdomyosarcoma and hepatocellular and renal cell carcinoma. In summary, our study provides evidence that perilipin 5 is not restricted to certain cell types but localizes to distinct lipid droplet subpopulations reflecting a possible function in oxidative energy supply in normal tissues and in diseases.
Collapse
Affiliation(s)
- Merita Hashani
- Department of General Pathology, Institute of Pathology, University Hospital, Heidelberg, Germany.,Institute of Pathology, University Hospital of Prishtina, Prishtina, Kosovo
| | - Hagen Roland Witzel
- Institute of Pathology, University Medicine, Johannes Gutenberg-University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Lena Maria Pawella
- Department of General Pathology, Institute of Pathology, University Hospital, Heidelberg, Germany
| | - Judith Lehmann-Koch
- Department of General Pathology, Institute of Pathology, University Hospital, Heidelberg, Germany
| | - Jens Schumacher
- Department of Internal Medicine III, Division of Translational and Experimental Oncology, University Hospital Mainz, Mainz, Germany
| | - Gunhild Mechtersheimer
- Department of General Pathology, Institute of Pathology, University Hospital, Heidelberg, Germany
| | - Martina Schnölzer
- Functional Proteome Analysis, German Cancer Research Center Heidelberg (DKFZ), INF 580, Heidelberg, Germany
| | - Peter Schirmacher
- Department of General Pathology, Institute of Pathology, University Hospital, Heidelberg, Germany
| | - Wilfried Roth
- Institute of Pathology, University Medicine, Johannes Gutenberg-University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Beate Katharina Straub
- Department of General Pathology, Institute of Pathology, University Hospital, Heidelberg, Germany. .,Institute of Pathology, University Medicine, Johannes Gutenberg-University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany.
| |
Collapse
|
47
|
Xu S, Zhang X, Liu P. Lipid droplet proteins and metabolic diseases. Biochim Biophys Acta Mol Basis Dis 2018; 1864:1968-1983. [DOI: 10.1016/j.bbadis.2017.07.019] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 07/14/2017] [Accepted: 07/19/2017] [Indexed: 12/13/2022]
|
48
|
Zhang P, Meng L, Song L, Du J, Du S, Cui W, Liu C, Li F. Roles of Perilipins in Diseases and Cancers. Curr Genomics 2018; 19:247-257. [PMID: 29755288 PMCID: PMC5930447 DOI: 10.2174/1389202918666170915155948] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 01/03/2017] [Accepted: 04/25/2017] [Indexed: 12/14/2022] Open
Abstract
Perilipins, an ancient family of lipid droplet-associated proteins, are embedded in a phospho-lipid monolayer of intracellular lipid droplets. The core of lipid droplets is composed of neutral fat, which mainly includes triglyceride and cholesterol ester. Perilipins are closely related to the function of lipid droplets, and they mediate lipid metabolism and storage. Therefore, perilipins play an important role in the development of obesity, diabetes, cancer, hepatic diseases, atherosclerosis, and carcinoma, which are caused by abnormal lipid metabolism. Accumulation of lipid droplets is a common phenomenon in tumor cells. Available data on the pathophysiology of perilipins and the relationship of perilipins with endocrine metabolic diseases and cancers are summarized in this mini-review. The research progress on this family offers novel insights into the therapeutic strategies for these diseases.
Collapse
Affiliation(s)
- Pengpeng Zhang
- Department of Pathology and The Key Laboratories for Xinjiang Endemic and Ethnic Diseases, School of Medicine, Shihezi University, Shihezi 832002, Xinjiang, China
| | - Lian Meng
- Department of Pathology and The Key Laboratories for Xinjiang Endemic and Ethnic Diseases, School of Medicine, Shihezi University, Shihezi 832002, Xinjiang, China
| | - Lingxie Song
- Department of Pathology and The Key Laboratories for Xinjiang Endemic and Ethnic Diseases, School of Medicine, Shihezi University, Shihezi 832002, Xinjiang, China
| | - Juan Du
- Department of Pathology and The Key Laboratories for Xinjiang Endemic and Ethnic Diseases, School of Medicine, Shihezi University, Shihezi 832002, Xinjiang, China
| | - Shutong Du
- Department of Pathology and The Key Laboratories for Xinjiang Endemic and Ethnic Diseases, School of Medicine, Shihezi University, Shihezi 832002, Xinjiang, China
| | - Wenwen Cui
- Department of Pathology and The Key Laboratories for Xinjiang Endemic and Ethnic Diseases, School of Medicine, Shihezi University, Shihezi 832002, Xinjiang, China
| | - Chunxia Liu
- Department of Pathology and The Key Laboratories for Xinjiang Endemic and Ethnic Diseases, School of Medicine, Shihezi University, Shihezi 832002, Xinjiang, China
| | - Feng Li
- Department of Pathology and The Key Laboratories for Xinjiang Endemic and Ethnic Diseases, School of Medicine, Shihezi University, Shihezi 832002, Xinjiang, China.,Department of Pathology, Beijing Chaoyang Hospital, Capital Medical University, Beijing100020, China
| |
Collapse
|
49
|
Campana B, Calabrese D, Matter MS, Terracciano LM, Wieland SF, Heim MH. In vivo analysis at the cellular level reveals similar steatosis induction in both hepatitis C virus genotype 1 and 3 infections. J Viral Hepat 2018; 25:262-271. [PMID: 29086446 DOI: 10.1111/jvh.12816] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 09/26/2017] [Indexed: 12/12/2022]
Abstract
Steatosis is a frequent histological feature of hepatitis C virus (HCV) infection. Cohort studies of patients with chronic hepatitis C identified HCV genotype 3 (HCV GT3) as the prevalent steatotic genotype. Moreover, Huh-7 cells over-expressing HCV GT3 core protein accumulate more triglyceride in larger lipid droplets than cells expressing core proteins of other HCV genotypes. However, little is known about the relationship of steatosis and HCV infection at the cellular level in vivo. In this study, we used highly sensitive multiplex in situ hybridization methodology together with lipid staining to investigate HCV-induced lipid droplet accumulation at the cellular level in liver biopsies. Consistent with previous reports, histological steatosis grades were significantly higher in GT3 compared to GT1 infected livers, but independent of viral load. Using nile red lipid stainings, we observed that the frequency of lipid droplet containing cells was similar in HCV GT1- and HCV GT3-infected livers. Lipid droplet formation preferentially occurred in HCV-infected cells irrespective of the genotype, but was also observed in noninfected cells. These findings demonstrate that the main difference between GT1- and GT3-induced steatosis is the size of lipid droplets, but not the number or relative distribution of lipid droplets in infected vs uninfected hepatocytes.
Collapse
Affiliation(s)
- B Campana
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland.,Division of Gastroenterology and Hepatology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - D Calabrese
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - M S Matter
- Institute of Pathology, Molecular Pathology Division, University Hospital of Basel, University of Basel, Basel, Switzerland
| | - L M Terracciano
- Institute of Pathology, Molecular Pathology Division, University Hospital of Basel, University of Basel, Basel, Switzerland
| | - S F Wieland
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - M H Heim
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland.,Division of Gastroenterology and Hepatology, University Hospital Basel, University of Basel, Basel, Switzerland
| |
Collapse
|
50
|
|