1
|
Slarve I, Wang Y, Ding Y, Niu X, Tang Q, Jia C, Tu T, Hong H, Zhang G, Gu Y, Xu Z, Skinner S, He L, Hua B, Nguyen P, Zhou Y, Chen L, Ashouri K, Martynova A, Nakhoul C, Rastegarpour A, Alachkar H, Lenz HJ, El-Khoueiry A, Sher L, Chopra S, Yuan L, Stiles BL. Isoform specific regulation of osteopontin by AKT2 in hepatocytes and livers. Cell Signal 2025; 132:111799. [PMID: 40216173 DOI: 10.1016/j.cellsig.2025.111799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2025] [Accepted: 04/08/2025] [Indexed: 04/18/2025]
Abstract
Elevated levels of osteopontin (OPN), an inflammatory cytokine, are correlated with chronic inflammatory conditions and liver cancer. In this study, we explored the regulation of OPN in liver and hepatocytes by AKT1 vs. AKT2, the two AKT isoforms expressed in hepatocytes and livers. Using a mouse model lacking PTEN (phosphatase and tensin homologue deleted on chromosome 10), the negative regulator of phosphatidylinositol 3-kinase (PI3K)/AKT signaling, expression of secreted phosphoprotein 1 (Spp1), the gene encoding OPN, was found to be the topmost significantly upregulated gene in the liver. Using an add-back experiment in hepatocytes isolated from these mice, we show that PTEN regulates the expression of Spp1 mRNA as well as OPN protein levels. Exploring how PTEN regulates the expression of Spp1/OPN, we investigated the differential roles of AKT1 vs. AKT2 using hepatocytes isolated from mice lacking each AKT isoform in the liver. We showed here that levels of Spp1/OPN in hepatocytes are lost with deletion of Akt2 but not Akt1. Deletion of Akt2 significantly attenuated both basal expression of OPN and its response to IGF-1 stimulation. AKT1 loss, on the other hand, permitted more robust induction of OPN by IGF-1 stimulation. Furthermore, mice lacking AKT2 and PTEN exhibit significantly lower OPN expression in the liver. Together, this study showed that OPN levels are regulated by the PI3K/AKT signal in hepatocytes and that AKT2 but not AKT1 is responsible for its induction in response to stimulation of the PI3K signaling pathway.
Collapse
Affiliation(s)
- Ielyzaveta Slarve
- Pharmacology and Pharmaceutical Sciences, Mann School of Pharmacy, University of Southern California, Los Angeles, CA 90033, USA
| | - Yushan Wang
- Pharmacology and Pharmaceutical Sciences, Mann School of Pharmacy, University of Southern California, Los Angeles, CA 90033, USA
| | - Yining Ding
- Pharmacology and Pharmaceutical Sciences, Mann School of Pharmacy, University of Southern California, Los Angeles, CA 90033, USA
| | - Xiaoteng Niu
- Pharmacology and Pharmaceutical Sciences, Mann School of Pharmacy, University of Southern California, Los Angeles, CA 90033, USA
| | - Qi Tang
- Pharmacology and Pharmaceutical Sciences, Mann School of Pharmacy, University of Southern California, Los Angeles, CA 90033, USA
| | - Chengyou Jia
- Pharmacology and Pharmaceutical Sciences, Mann School of Pharmacy, University of Southern California, Los Angeles, CA 90033, USA; Department of Nuclear Medicine, Central Laboratory for Medical Research, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Taojian Tu
- Pharmacology and Pharmaceutical Sciences, Mann School of Pharmacy, University of Southern California, Los Angeles, CA 90033, USA
| | - Handan Hong
- Pharmacology and Pharmaceutical Sciences, Mann School of Pharmacy, University of Southern California, Los Angeles, CA 90033, USA
| | - Guo Zhang
- Pharmacology and Pharmaceutical Sciences, Mann School of Pharmacy, University of Southern California, Los Angeles, CA 90033, USA
| | - Yiwei Gu
- Pharmacology and Pharmaceutical Sciences, Mann School of Pharmacy, University of Southern California, Los Angeles, CA 90033, USA
| | - Zifei Xu
- Pharmacology and Pharmaceutical Sciences, Mann School of Pharmacy, University of Southern California, Los Angeles, CA 90033, USA
| | - Samantha Skinner
- Pharmacology and Pharmaceutical Sciences, Mann School of Pharmacy, University of Southern California, Los Angeles, CA 90033, USA
| | - Lina He
- Pharmacology and Pharmaceutical Sciences, Mann School of Pharmacy, University of Southern California, Los Angeles, CA 90033, USA
| | - Brittney Hua
- Pharmacology and Pharmaceutical Sciences, Mann School of Pharmacy, University of Southern California, Los Angeles, CA 90033, USA
| | - Phillip Nguyen
- Pharmacology and Pharmaceutical Sciences, Mann School of Pharmacy, University of Southern California, Los Angeles, CA 90033, USA
| | - Yiren Zhou
- Pharmacology and Pharmaceutical Sciences, Mann School of Pharmacy, University of Southern California, Los Angeles, CA 90033, USA
| | - Lulu Chen
- Pharmacology and Pharmaceutical Sciences, Mann School of Pharmacy, University of Southern California, Los Angeles, CA 90033, USA
| | - Karam Ashouri
- Division of Medical Oncology, Keck School of Medicine, University of Southern California, Norris Comprehensive Cancer Center, Los Angeles, CA 90033, USA
| | - Anastasia Martynova
- Division of Medical Oncology, Keck School of Medicine, University of Southern California, Norris Comprehensive Cancer Center, Los Angeles, CA 90033, USA
| | - Christina Nakhoul
- Division of Medical Oncology, Keck School of Medicine, University of Southern California, Norris Comprehensive Cancer Center, Los Angeles, CA 90033, USA
| | - Ali Rastegarpour
- Division of Medical Oncology, Keck School of Medicine, University of Southern California, Norris Comprehensive Cancer Center, Los Angeles, CA 90033, USA
| | - Houda Alachkar
- Department of Clinical Pharmacy, Mann School of Pharmacy, University of Southern California, Los Angeles, CA 90089, USA
| | - Heinz-Josef Lenz
- Division of Medical Oncology, Keck School of Medicine, University of Southern California, Norris Comprehensive Cancer Center, Los Angeles, CA 90033, USA
| | - Anthony El-Khoueiry
- Division of Medical Oncology, Keck School of Medicine, University of Southern California, Norris Comprehensive Cancer Center, Los Angeles, CA 90033, USA
| | - Linda Sher
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Shefali Chopra
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Liyun Yuan
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Bangyan Li Stiles
- Pharmacology and Pharmaceutical Sciences, Mann School of Pharmacy, University of Southern California, Los Angeles, CA 90033, USA; Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| |
Collapse
|
2
|
Tang BX, Zhang Y, Sun DD, Liu QY, Li C, Wang PP, Gao LX, Zhang XM, Li J, Zhu WL, Zang Y. Luteolin-7-diglucuronide, a novel PTP1B inhibitor, ameliorates hepatic stellate cell activation and liver fibrosis in mice. Acta Pharmacol Sin 2025; 46:122-133. [PMID: 39103531 PMCID: PMC11697251 DOI: 10.1038/s41401-024-01351-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 07/02/2024] [Indexed: 08/07/2024]
Abstract
Liver fibrosis, one of the leading causes of morbidity and mortality worldwide, lacks effective therapy. The activation of hepatic stellate cells (HSCs) is the dominant event in hepatic fibrogenesis. Luteolin-7-diglucuronide (L7DG) is the major flavonoid extracted from Perilla frutescens and Verbena officinalis. Their beneficial effects in the treatment of liver diseases were well documented. In this study we investigated the anti-fibrotic activities of L7DG and the potential mechanisms. We established TGF-β1-activated mouse primary hepatic stellate cells (pHSCs) and human HSC line LX-2 as in vitro liver fibrosis models. Co-treatment with L7DG (5, 20, 50 μM) dose-dependently decreased TGF-β1-induced expression of fibrotic markers collagen 1, α-SMA and fibronectin. In liver fibrosis mouse models induced by CCl4 challenge alone or in combination with HFHC diet, administration of L7DG (40, 150 mg·kg-1·d-1, i.g., for 4 or 8 weeks) dose-dependently attenuated hepatic histopathological injury and collagen accumulation, decreased expression of fibrogenic genes. By conducting target prediction, molecular docking and enzyme activity detection, we identified L7DG as a potent inhibitor of protein tyrosine phosphatase 1B (PTP1B) with an IC50 value of 2.10 µM. Further studies revealed that L7DG inhibited PTP1B activity, up-regulated AMPK phosphorylation and subsequently inhibited HSC activation. This study demonstrates that the phytochemical L7DG may be a potential therapeutic candidate for the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Bi-Xi Tang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, 201203, China
- Stake Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yong Zhang
- Stake Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Dan-Dan Sun
- Stake Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Qin-Yi Liu
- Stake Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Cong Li
- Stake Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Pei-Pei Wang
- Stake Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Li-Xin Gao
- Stake Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xue-Mei Zhang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, 201203, China.
| | - Jia Li
- Stake Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, 100049, Beijing, China.
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264117, China.
| | - Wei-Liang Zhu
- Stake Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| | - Yi Zang
- Lingang Laboratory, Shanghai, 201203, China.
| |
Collapse
|
3
|
Nazeer B, Khawar MB, Khalid MU, Hamid SE, Rafiq M, Abbasi MH, Sheikh N, Ali A, Fatima H, Ahmad S. Emerging role of lipophagy in liver disorders. Mol Cell Biochem 2024; 479:1-11. [PMID: 36943663 DOI: 10.1007/s11010-023-04707-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 03/10/2023] [Indexed: 03/23/2023]
Abstract
Lipophagy is a selective degradation of lipids by a lysosomal-mediated pathway, and dysregulation of lipophagy is linked with the pathological hallmark of many liver diseases. Downregulation of lipophagy in liver cells results in abnormal accumulation of LDs (Lipid droplets) in hepatocytes which is a characteristic feature of several liver pathologies such as nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH). Contrarily, upregulation of lipophagy in activated hepatic stellate cells (HSCs) is associated with hepatic fibrosis and cirrhosis. Lipid metabolism reprogramming in violent cancer cells contributes to the progression of liver cancer. In this review, we have summarized the recent studies focusing on various components of the lipophagic machinery that can be modulated for their potential role as therapeutic agents against a wide range of liver diseases.
Collapse
Affiliation(s)
- Bismillah Nazeer
- Molecular Medicine and Cancer Therapeutics Lab, Department of Zoology, Faculty of Sciences, University of Central Punjab, Lahore, Pakistan
| | - Muhammad Babar Khawar
- Applied Molecular Biology and Biomedicine Lab, Department of Zoology, University of Narowal, Narowal, Pakistan.
| | - Muhammad Usman Khalid
- Molecular Medicine and Cancer Therapeutics Lab, Department of Zoology, Faculty of Sciences, University of Central Punjab, Lahore, Pakistan
| | - Syeda Eisha Hamid
- Molecular Medicine and Cancer Therapeutics Lab, Department of Zoology, Faculty of Sciences, University of Central Punjab, Lahore, Pakistan
| | - Mussarat Rafiq
- Cell and Molecular Biology Lab, Institute of Zoology, University of the Punjab, Lahore, Pakistan
| | | | - Nadeem Sheikh
- Cell and Molecular Biology Lab, Institute of Zoology, University of the Punjab, Lahore, Pakistan.
| | - Ahmad Ali
- Molecular Medicine and Cancer Therapeutics Lab, Department of Zoology, Faculty of Sciences, University of Central Punjab, Lahore, Pakistan
| | - Hooriya Fatima
- Molecular Medicine and Cancer Therapeutics Lab, Department of Zoology, Faculty of Sciences, University of Central Punjab, Lahore, Pakistan
| | - Sadia Ahmad
- Molecular Medicine and Cancer Therapeutics Lab, Department of Zoology, Faculty of Sciences, University of Central Punjab, Lahore, Pakistan
| |
Collapse
|
4
|
Mao FF, Gao SS, Huang YJ, Zhou N, Feng JK, Liu ZH, Zhang YQ, Yuan LY, Wei G, Cheng SQ. Network pharmacology-based analysis of Resinacein S against non-alcoholic fatty liver disease by modulating lipid metabolism. Front Nutr 2023; 10:1076569. [PMID: 36866057 PMCID: PMC9971728 DOI: 10.3389/fnut.2023.1076569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 01/11/2023] [Indexed: 02/16/2023] Open
Abstract
Background Ganoderma lucidum is reportedly the best source of traditional natural bioactive constituents. Ganoderma triterpenoids (GTs) have been verified as an alternative adjuvant for treating leukemia, cancer, hepatitis and diabetes. One of the major triterpenoids, Resinacein S, has been found to regulate lipid metabolism and mitochondrial biogenesis. Nonalcoholic fatty liver disease (NAFLD) is a common chronic liver disease that has become a major public health problem. Given the regulatory effects on lipid metabolism of Resinacein S, we sought to explore potential protective effects against NAFLD. Methods Resinacein S was extracted and isolated from G. lucidum. And mice were fed with high fat diet with or without Resinacein S to detect hepatic steatosis. According to Network Pharmacology and RNA-seq, we analyzed the hub genes of Resinacein S against NAFLD disease. Results Our results can be summarized as follows: (1) The structure of Resinacein S was elucidated using NMR and MS methods. (2) Resinacein S treatment could significantly attenuate high-fat diet (HFD)-induced hepatic steatosis and hepatic lipid accumulation in mouse. (3) GO terms, KEGG pathways and the PPI network of Resinacein S induced Differentially Expressed Genes (DEGs) demonstrated the key target genes of Resinacein S against NAFLD. (4) The hub proteins in PPI network analysis could be used for NAFLD diagnosis and treatment as drug targets. Conclusion Resinacein S can significantly change the lipid metabolism in liver cells and yield a protective effect against steatosis and liver injury. Intersected proteins between NAFLD related genes and Resinacein S-induced DEGs, especially the hub protein in PPI network analysis, can be used to characterize targets of Resinacein S against NAFLD.
Collapse
Affiliation(s)
- Fei-Fei Mao
- Tongji University Cancer Center, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Shan-Shan Gao
- Tongji University Cancer Center, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yan-Jie Huang
- Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Nian Zhou
- Tongji University Cancer Center, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jin-Kai Feng
- Department of Hepatic Surgery VI, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Zong-Han Liu
- Department of Hepatic Surgery VI, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Yu-Qing Zhang
- Cancer Center, Yue Yang Hospital of Integrative Traditional Chinese and Western Medicine, Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lu-Yun Yuan
- Cancer Center, Yue Yang Hospital of Integrative Traditional Chinese and Western Medicine, Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Gang Wei
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, China,*Correspondence: Gang Wei, ✉
| | - Shu-Qun Cheng
- Tongji University Cancer Center, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China,Department of Hepatic Surgery VI, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China,Shuqun Cheng, ✉
| |
Collapse
|
5
|
Alba MM, Ebright B, Hua B, Slarve I, Zhou Y, Jia Y, Louie SG, Stiles BL. Eicosanoids and other oxylipins in liver injury, inflammation and liver cancer development. Front Physiol 2023; 14:1098467. [PMID: 36818443 PMCID: PMC9932286 DOI: 10.3389/fphys.2023.1098467] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 01/16/2023] [Indexed: 02/05/2023] Open
Abstract
Liver cancer is a malignancy developed from underlying liver disease that encompasses liver injury and metabolic disorders. The progression from these underlying liver disease to cancer is accompanied by chronic inflammatory conditions in which liver macrophages play important roles in orchestrating the inflammatory response. During this process, bioactive lipids produced by hepatocytes and macrophages mediate the inflammatory responses by acting as pro-inflammatory factors, as well as, playing roles in the resolution of inflammation conditions. Here, we review the literature discussing the roles of bioactive lipids in acute and chronic hepatic inflammation and progression to cancer.
Collapse
Affiliation(s)
- Mario M. Alba
- Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, Unites States
| | - Brandon Ebright
- Clinical Pharmacy, School of Pharmacy, University of Southern California, Los Angeles, CA, Unites States
| | - Brittney Hua
- Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, Unites States
| | - Ielyzaveta Slarve
- Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, Unites States
| | - Yiren Zhou
- Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, Unites States
| | - Yunyi Jia
- Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, Unites States
| | - Stan G. Louie
- Clinical Pharmacy, School of Pharmacy, University of Southern California, Los Angeles, CA, Unites States
| | - Bangyan L. Stiles
- Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, Unites States
- Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, Unites States
| |
Collapse
|
6
|
Fornari F, Giovannini C, Piscaglia F, Gramantieri L. Animal Models of Hepatocellular Carcinoma: Current Applications in Clinical Research. J Hepatocell Carcinoma 2022; 9:1263-1278. [PMID: 36523954 PMCID: PMC9744868 DOI: 10.2147/jhc.s347946] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 11/23/2022] [Indexed: 01/11/2025] Open
Abstract
In the last decade, relevant advances have occurred in the treatment of hepatocellular carcinoma (HCC), with novel drugs entering the clinical practice, among which tyrosine kinase inhibitors (TKIs) such as lenvatinib, cabozantinib and regorafenib, and immune checkpoint inhibitors (ICPIs) either alone or in combination with VEGF inhibitors. Clinical trials have driven the introduction of such novel molecules into the clinics but, at present, no biomarker drives the choice of first-line options, which relies only upon clinical and imaging assessment. Remarkably, clinical and imaging-based evaluations do not consider the huge heterogeneity of HCC and do not allow to realize the potential of personalized treatments. Preclinical research still does not inform the design of clinical trials, even though many animal models mimicking specific subgroups of HCC are available and might provide relevant information. Although animal models directly informing the clinical practice, such as patients-derived xenografts, are not used to help the choice of treatment in advanced HCC, however, the preclinical research can count on a wide range of valuable models. Here we will review some HCC models which might turn informative for specific questions in defined patient subgroups, and we will describe recent preclinical studies for the mechanistic evaluation of immunotherapy-based treatment approaches. To this aim, we will mainly focus on two issues: (i) HCC models informative on NAFLD-NASH HCC and (ii) HCC models helping to elucidate mechanisms underneath immunotherapy. We have chosen these two settings since they represent, respectively, the most rapidly arising cause of chronic liver disease (CLD) and HCC in western countries and the most promising therapeutic option for advanced HCC.
Collapse
Affiliation(s)
- Francesca Fornari
- Centre for Applied Biomedical Research - CRBA, University of Bologna, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
- Department for Life Quality Studies, University of Bologna, Rimini, Italy
| | - Catia Giovannini
- Centre for Applied Biomedical Research - CRBA, University of Bologna, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Fabio Piscaglia
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Laura Gramantieri
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
| |
Collapse
|
7
|
Tu T, Alba MM, Datta AA, Hong H, Hua B, Jia Y, Khan J, Nguyen P, Niu X, Pammidimukkala P, Slarve I, Tang Q, Xu C, Zhou Y, Stiles BL. Hepatic macrophage mediated immune response in liver steatosis driven carcinogenesis. Front Oncol 2022; 12:958696. [PMID: 36276076 PMCID: PMC9581256 DOI: 10.3389/fonc.2022.958696] [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: 05/31/2022] [Accepted: 08/17/2022] [Indexed: 12/02/2022] Open
Abstract
Obesity confers an independent risk for carcinogenesis. Classically viewed as a genetic disease, owing to the discovery of tumor suppressors and oncogenes, genetic events alone are not sufficient to explain the progression and development of cancers. Tumor development is often associated with metabolic and immunological changes. In particular, obesity is found to significantly increase the mortality rate of liver cancer. As its role is not defined, a fundamental question is whether and how metabolic changes drive the development of cancer. In this review, we will dissect the current literature demonstrating that liver lipid dysfunction is a critical component driving the progression of cancer. We will discuss the involvement of inflammation in lipid dysfunction driven liver cancer development with a focus on the involvement of liver macrophages. We will first discuss the association of steatosis with liver cancer. This will be followed with a literature summary demonstrating the importance of inflammation and particularly macrophages in the progression of liver steatosis and highlighting the evidence that macrophages and macrophage produced inflammatory mediators are critical for liver cancer development. We will then discuss the specific inflammatory mediators and their roles in steatosis driven liver cancer development. Finally, we will summarize the molecular pattern (PAMP and DAMP) as well as lipid particle signals that are involved in the activation, infiltration and reprogramming of liver macrophages. We will also discuss some of the therapies that may interfere with lipid metabolism and also affect liver cancer development.
Collapse
Affiliation(s)
- Taojian Tu
- Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, United States
| | - Mario M. Alba
- Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, United States
| | - Aditi A. Datta
- Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, United States
| | - Handan Hong
- Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, United States
| | - Brittney Hua
- Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, United States
| | - Yunyi Jia
- Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, United States
| | - Jared Khan
- Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, United States
| | - Phillip Nguyen
- Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, United States
| | - Xiatoeng Niu
- Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, United States
| | - Pranav Pammidimukkala
- Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, United States
| | - Ielyzaveta Slarve
- Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, United States
| | - Qi Tang
- Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, United States
| | - Chenxi Xu
- Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, United States
| | - Yiren Zhou
- Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, United States
| | - Bangyan L. Stiles
- Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, United States
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- *Correspondence: Bangyan L. Stiles,
| |
Collapse
|
8
|
Jie L, Hong RT, Zhang YJ, Sha LL, Chen W, Ren XF. Melatonin Alleviates Liver Fibrosis by Inhibiting Autophagy. Curr Med Sci 2022; 42:498-504. [PMID: 35583587 DOI: 10.1007/s11596-022-2530-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 06/18/2021] [Indexed: 11/09/2022]
Abstract
OBJECTIVE Melatonin has been reported to suppress inflammation and alleviate liver fibrosis, but its effect on autophagy in liver fibrosis has not been studied. This study investigated the effect of melatonin on autophagy in an animal model of liver fibrosis and the hepatic stellate cell (HSC)-T6 cell line. METHODS The model was established in rats through carbon tetrachloride treatment, and melatonin was administered at three doses (2.5, 5.0, and 10.0 mg/kg). Haematoxylin and eosin staining and Van Gieson's staining were performed to examine the pathological changes of liver. The expression of alpha-smooth muscle actin (α-SMA) and Beclin1 in liver tissues was detected by immunohistochemical staining. The protein levels of α-SMA, Beclin1 and LC3 in the animal model were detected by Western blot analysis, and gene levels of Beclin1 and LC3 were detected by quantitative real-time PCR (qRT-PCR) in the animal model. HSC-T6 cells were activated by platelet-derived growth factor-BB (PDGF-BB). The expression of α-SMA, Beclin1 and collagen I was detected by Western blot analysis, and the gene expression of Beclin1 and LC3 was detected by qRT-PCR. RESULTS Melatonin reduced the expression of α-SMA, Beclin1 and LC3 in liver tissues. In addition, melatonin inhibited the activation of HSC-T6 cells and the expression of α-SMA, Beclin1 and LC3 in these cells. These results revealed that melatonin could inhibit autophagy and HSC activation. CONCLUSION Melatonin might ameliorate liver fibrosis by regulating autophagy, suggesting that melatonin is a potential therapeutic agent for liver fibrosis.
Collapse
Affiliation(s)
- Lei Jie
- Department of Gastroenterology, First Affiliated Hospital of Anhui Medical University, Key Laboratory of Digestive Disease of Anhui Province, Hefei, 230022, China
| | - Ru-Tao Hong
- Department of Gastroenterology, First Affiliated Hospital of Anhui Medical University, Key Laboratory of Digestive Disease of Anhui Province, Hefei, 230022, China.
| | - Yu-Jie Zhang
- Department of Gastroenterology, First Affiliated Hospital of Anhui Medical University, Key Laboratory of Digestive Disease of Anhui Province, Hefei, 230022, China
| | - Lu-Lin Sha
- Department of Critical Care Medicine of Cardiothoracic Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Wei Chen
- Department of Gastroenterology, First Affiliated Hospital of Anhui Medical University, Key Laboratory of Digestive Disease of Anhui Province, Hefei, 230022, China
| | - Xiao-Fei Ren
- Department of Gastroenterology, First Affiliated Hospital of Anhui Medical University, Key Laboratory of Digestive Disease of Anhui Province, Hefei, 230022, China
| |
Collapse
|
9
|
Ma L, Wei J, Zeng Y, Liu J, Xiao E, Kang Y, Kang Y. Mesenchymal stem cell-originated exosomal circDIDO1 suppresses hepatic stellate cell activation by miR-141-3p/PTEN/AKT pathway in human liver fibrosis. Drug Deliv 2022; 29:440-453. [PMID: 35099348 PMCID: PMC8812765 DOI: 10.1080/10717544.2022.2030428] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Liver fibrosis is a common pathologic stage of the development of liver failure. It has showed that exosomes loaded with therapeutic circRNAs can be manufactured in bulk by exosome secreted cells in vitro, thus enabling personalized treatment. This study aimed to investigate the role of exosome-based delivery of circDIDO1 in liver fibrosis. Levels of genes and proteins were examined by qRT-PCR and Western blot. Cell proliferation, apoptosis, and cell cycle were analyzed by using cell counting kit-8 (CCK-8) assay, EdU assay, and flow cytometry, respectively. The binding between circDIDO1 and miR-141-3p was confirmed by dual-luciferase reporter, RNA pull-down and RIP assays. Exosomes were isolated by ultracentrifugation, and qualified by transmission electron microscopy (TEM), nanoparticle tracking analysis (NTA) and Western blot. CircDIDO1 overexpression or miR-141-3p inhibition suppressed the proliferation, reduced pro-fibrotic markers, and induced apoptosis as well as cell cycle arrest in hepatic stellate cells (HSCs) by blocking PTEN/AKT pathway. Mechanistically, circDIDO1 acted as an endogenous sponge for miR-141-3p, further rescue experiments showed that circDIDO1 suppressed HSC activation by targeting miR-141-3p. Extracellular circDIDO1 could be incorporated into exosomes isolated from mesenchymal stem cells (MSCs), and transmitted to HSCs to restrain HSC activation. Clinically, low levels of serum circDIDO1 in exosome were correlated with liver failure, and serum exosomal circDIDO1 had a well diagnostic value for liver fibrosis in liver failure patients. Transfer of circDIDO1 mediated by MSC-isolated exosomes suppressed HSC activation through the miR-141-3p/PTEN/AKT pathway, gaining a new insight into the prevention of liver fibrosis in liver failure patients.
Collapse
Affiliation(s)
- Li Ma
- Department of Infectious Diseases, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, China
| | - Junfeng Wei
- Department of Infectious Diseases, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, China
| | - Yanli Zeng
- Department of Infectious Diseases, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, China
| | - Junping Liu
- Department of Infectious Diseases, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, China
| | - Erhui Xiao
- Department of Infectious Diseases, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, China
| | - Yuehua Kang
- Department of Infectious Diseases, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, China
| | - Yi Kang
- Department of Infectious Diseases, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, China
| |
Collapse
|
10
|
Aggarwal R, Peng Z, Zeng N, Silva J, He L, Chen J, Debebe A, Tu T, Alba M, Chen CY, Stiles EX, Hong H, Stiles BL. Chronic Exposure to Palmitic Acid Down-Regulates AKT in Beta-Cells through Activation of mTOR. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:130-145. [PMID: 34619135 PMCID: PMC8759041 DOI: 10.1016/j.ajpath.2021.09.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 09/09/2021] [Accepted: 09/22/2021] [Indexed: 01/03/2023]
Abstract
High circulating lipids occurring in obese individuals and insulin-resistant patients are considered a contributing factor to type 2 diabetes. Exposure to high lipid concentration is proposed to both protect and damage beta-cells under different circumstances. Here, by feeding mice a high-fat diet (HFD) for 2 weeks to up to 14 months, the study showed that HFD initially causes the beta-cells to expand in population, whereas long-term exposure to HFD is associated with failure of beta-cells and the inability of animals to respond to glucose challenge. To prevent the failure of beta-cells and the development of type 2 diabetes, the molecular mechanisms that underlie this biphasic response of beta-cells to lipid exposure were explored. Using palmitic acid (PA) in cultured beta-cells and islets, the study demonstrated that chronic exposure to lipids leads to reduced viability and inhibition of cell cycle progression concurrent with down-regulation of a pro-growth/survival kinase AKT, independent of glucose. This AKT down-regulation by PA is correlated with the induction of mTOR/S6K activity. Inhibiting mTOR activity with rapamycin induced Raptor and restored AKT activity, allowing beta-cells to gain proliferation capacity that was lost after HFD exposure. In summary, a novel mechanism in which lipid exposure may cause the dipole effects on beta-cell growth was elucidated, where mTOR acts as a lipid sensor. These mechanisms can be novel targets for future therapeutic developments.
Collapse
Affiliation(s)
- Richa Aggarwal
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Zhechu Peng
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Ni Zeng
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Joshua Silva
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Lina He
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Jingyu Chen
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Anketse Debebe
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Taojian Tu
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Mario Alba
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Chien-Yu Chen
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Eileen X. Stiles
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Handan Hong
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Bangyan L. Stiles
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California,Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, California,Address correspondence to Bangyan L. Stiles, Ph.D., Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90033.
| |
Collapse
|
11
|
Carson JP, Robinson MW, Ramm GA, Gobert GN. RNA sequencing of LX-2 cells treated with TGF-β1 identifies genes associated with hepatic stellate cell activation. Mol Biol Rep 2021; 48:7677-7688. [PMID: 34648138 PMCID: PMC8604886 DOI: 10.1007/s11033-021-06774-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 09/14/2021] [Indexed: 11/10/2022]
Abstract
Background Hepatic stellate cells (HSCs) are liver-resident myofibroblast precursors responsible for the production of collagen and maintenance of the hepatic extracellular matrix (ECM). As such, they are generally associated with fibrotic liver diseases. HSCs become “activated” in response to tissue damage or pathogen invasion, a process most commonly driven by transforming growth factor-β1 (TGF-β1). Despite this, the full extent of TGF-β1 signalling in these cells is poorly understood. Clarifying the range and diversity of this signalling will further improve our understanding of the process of HSC activation. Methods and results RNA sequencing was used to quantitate the transcriptomic changes induced in LX-2 cells, an activated human HSC line, following TGF-b1 treatment. In total, 5,258 genes were found to be significantly differentially expressed with a false discovery rate cut-off of < 0.1. The topmost deregulated of these genes included those with no currently characterised role in either HSC activation or fibrotic processes, including CIITA and SERPINB2. In silico analysis revealed the prominent signalling pathways downstream of TGF-β1 in LX-2 cells. Conclusions In this study, we describe the genes and signalling pathways significantly deregulated in LX-2 cells following TGF-β1 treatment. We identified several highly deregulated genes with no currently characterised role in HSC activation, which may represent novel mediators of fibrotic responses in HSCs or the liver macroenvironment. This work may be of use in the identification of new markers of liver fibrosis and could provide insight into prospective genes or pathways that might be targeted for the amelioration of fibrotic liver disease in the future.
Collapse
Affiliation(s)
- Jack P. Carson
- School of Biological Sciences, Queen’s University Belfast, 19 Chlorine Gardens, BT9 5DL Belfast, UK
| | - Mark W. Robinson
- School of Biological Sciences, Queen’s University Belfast, 19 Chlorine Gardens, BT9 5DL Belfast, UK
| | - Grant A. Ramm
- QIMR Berghofer Medical Research Institute, Royal Brisbane Hospital, Locked Bag 2000, QLD 4029 Brisbane, Australia
- Faculty of Medicine, The University of Queensland, Level 6, Oral Health Centre (Building), Herston Road, 4006 Herston, QLD Australia
| | - Geoffrey N. Gobert
- School of Biological Sciences, Queen’s University Belfast, 19 Chlorine Gardens, BT9 5DL Belfast, UK
| |
Collapse
|
12
|
Chen CY, Li Y, Zeng N, He L, Zhang X, Tu T, Tang Q, Alba M, Mir S, Stiles EX, Hong H, Cadenas E, Stolz AA, Li G, Stiles BL. Inhibition of Estrogen-Related Receptor α Blocks Liver Steatosis and Steatohepatitis and Attenuates Triglyceride Biosynthesis. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:1240-1254. [PMID: 33894178 PMCID: PMC8261472 DOI: 10.1016/j.ajpath.2021.04.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 03/08/2021] [Accepted: 04/12/2021] [Indexed: 01/02/2023]
Abstract
The estrogen-related receptor (ERR) family of orphan nuclear receptors are transcriptional activators for genes involved in mitochondrial bioenergetics and metabolism. The goal of this study was to explore the role of ERRα in lipid metabolism and the potential effect of inhibiting ERRα on the development of nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH). In the current study, three experimental mouse models: high-fat diet, high-carbohydrate diet, and a genetic model of hepatic insulin resistance where the liver hyperinsulinemia signal is mimicked via hepatic deletion of Pten (phosphatase and tensin homolog deleted on chromosome 10), the negative regulator of the insulin/phosphatidylinositol 3-kinase signaling pathway, were used. A recently developed small-molecule inhibitor for ERRα was used to demonstrate that inhibiting ERRα blocked NAFLD development induced by either high-carbohydrate diet or high-fat diet feeding. ERRα inhibition also diminished lipid accumulation and attenuated NASH development in the Pten null mice. Glycerolipid synthesis was discovered as an additional mechanism for ERRα-regulated NAFLD/NASH development and glycerophosphate acyltransferase 4 was identified as a novel transcriptional target of ERRα. In summary, these results establish ERRα as a major transcriptional regulator of lipid biosynthesis in addition to its characterized primary function as a regulator for mitochondrial function. This study recognizes ERRα as a potential target for NAFLD/NASH treatment and elucidates novel signaling pathways regulated by ERRα.
Collapse
Affiliation(s)
- Chien-Yu Chen
- Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Yang Li
- Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Ni Zeng
- Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Lina He
- Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Xinwen Zhang
- Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Taojian Tu
- Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Qi Tang
- Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Mario Alba
- Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Sabrina Mir
- Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Eileen X Stiles
- Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Handan Hong
- Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Enrique Cadenas
- Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California; Department of Biochemistry, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Andrew A Stolz
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Gang Li
- Faculty of Health Sciences, University of Macau, Macau
| | - Bangyan L Stiles
- Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California; Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, California.
| |
Collapse
|
13
|
Chen J, Debebe A, Zeng N, Kopp J, He L, Sander M, Stiles BL. Transformation of SOX9 + cells by Pten deletion synergizes with steatotic liver injury to drive development of hepatocellular and cholangiocarcinoma. Sci Rep 2021; 11:11823. [PMID: 34083580 PMCID: PMC8175600 DOI: 10.1038/s41598-021-90958-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 05/19/2021] [Indexed: 01/07/2023] Open
Abstract
SOX9 (Sex-determining region Y Box 9) is a well-characterized transcription factor that is a marker for progenitor cells in various tissues. In the liver, cells delineated by SOX9 are responsible for regenerating liver parenchyma when cell proliferation is impaired following chronic injury. However, whether these SOX9+ cells play a role in liver carcinogenesis has not been fully understood, although high SOX9 expression has been linked to poor survival outcome in liver cancer patients. To address this question, we developed a liver cancer mouse model (PtenloxP/loxP; Sox9-CreERT+; R26RYFP) where tumor suppressor Pten (phosphatase and tensin homolog deleted on chromosome ten) is deleted in SOX9+ cells following tamoxifen injection. In this paper, we employ lineage-tracing to demonstrate the tumorigenicity potential of the Pten-, SOX9+ cells. We show that these cells are capable of giving rise to mixed-lineage tumors that manifest features of both hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (CCA). Our results suggest that PTEN loss induces the transformation of SOX9+ cells. We further show that to activate these transformed SOX9+ cells, the presence of liver injury is crucial. Liver injury, induced by hepatotoxin 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC) or high-fat diet (HFD), substantially increases tumor incidence and accelerates liver carcinogenesis from SOX9+ cells in Pten null mice but not in control mice. We further examine the mechanisms underlying tumor formation in this model to show that concurrent with the induction of niche signal (i.e., Wnt signaling), liver injury significantly stimulates the expansion of tumor-initiating cells (TICs). Together, these data show that (1) SOX9+ cells have the potential to become TICs following the primary transformation (i.e. Pten deletion) and that (2) liver injury is necessary for promoting the activation and proliferation of transformed SOX9+ cells, resulting in the genesis of mixed-lineage liver tumors.
Collapse
Affiliation(s)
- Jingyu Chen
- Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, 90033, USA
| | - Anketse Debebe
- Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, 90033, USA
| | - Ni Zeng
- Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, 90033, USA
| | - Janel Kopp
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Lina He
- Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, 90033, USA
| | - Maike Sander
- Department of Pediatrics and Cellar and Molecular Medicine, UCSD, La Jolla, CA, 92093, USA
| | - Bangyan L Stiles
- Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, 90033, USA.
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA.
| |
Collapse
|
14
|
Riaz F, Chen Q, Lu K, Osoro EK, Wu L, Feng L, Zhao R, Yang L, Zhou Y, He Y, Zhu L, Du X, Sadiq M, Yang X, Li D. Inhibition of miR-188-5p alleviates hepatic fibrosis by significantly reducing the activation and proliferation of HSCs through PTEN/PI3K/AKT pathway. J Cell Mol Med 2021; 25:4073-4087. [PMID: 33689215 PMCID: PMC8051718 DOI: 10.1111/jcmm.16376] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 01/29/2021] [Accepted: 02/03/2021] [Indexed: 02/06/2023] Open
Abstract
Persistent hepatic damage and chronic inflammation in liver activate the quiescent hepatic stellate cells (HSCs) and cause hepatic fibrosis (HF). Several microRNAs regulate the activation and proliferation of HSCs, thereby playing a critical role in HF progression. Previous studies have reported that miR‐188‐5p is dysregulated during the process of HF. However, the role of miR‐188‐5p in HF remains unclear. This study investigated the potential role of miR‐188‐5p in HSCs and HF. Firstly, we validated the miR‐188‐5p expression in primary cells isolated from liver of carbon tetrachloride (CCl4)‐induced mice, TGF‐β1‐induced LX‐2 cells, livers from 6‐month high‐fat diet (HFD)‐induced rat and 4‐month HFD‐induced mice NASH models, and human non‐alcoholic fatty liver disease (NAFLD) patients. Furthermore, we used miR‐188‐5p inhibitors to investigate the therapeutic effects of miR‐188‐5p inhibition in the HFD + CCl4 induced in vivo model and the potential role of miR‐188‐5p in the activation and proliferation of HSCs. This present study reported that miR‐188‐5p expression is significantly increased in the human NAFLD, HSCs isolated from liver of CCl4 induced mice, and in vitro and in vivo models of HF. Mimicking the miR‐188‐5p resulted in the up‐regulation of HSC activation and proliferation by directly targeting the phosphatase and tensin homolog (PTEN). Moreover, inhibition of miR‐188‐5p reduced the activation and proliferation markers of HSCs through PTEN/AKT pathway. Additionally, in vivo inhibition of miR‐188‐5p suppressed the HF parameters, pro‐fibrotic and pro‐inflammatory genes, and fibrosis. Collectively, our results uncover the pro‐fibrotic role of miR‐188‐5p. Furthermore, we demonstrated that miR‐188‐5p inhibition decreases the severity of HF by reducing the activation and proliferation of HSCs through PTEN/AKT pathway.
Collapse
Affiliation(s)
- Farooq Riaz
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Qian Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Kaikai Lu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Ezra Kombo Osoro
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Litao Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Lina Feng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Rong Zhao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Luyun Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Yimeng Zhou
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Yingli He
- Department of Infectious Diseases, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Li Zhu
- Department of Infectious Diseases, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaojuan Du
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Muhammad Sadiq
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Xudong Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| | - Dongmin Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, China
| |
Collapse
|
15
|
Optimization of the isolation procedure and culturing conditions for hepatic stellate cells obtained from mouse. Biosci Rep 2021; 41:227415. [PMID: 33350435 PMCID: PMC7823195 DOI: 10.1042/bsr20202514] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 11/28/2020] [Accepted: 12/18/2020] [Indexed: 12/28/2022] Open
Abstract
Liver fibrosis (LF) mortality rate is approximately 2 million per year. Irrespective of the etiology of LF, a key element in its development is the transition of hepatic stellate cells (HSCs) from a quiescent phenotype to a myofibroblast-like cell with the production of fibrotic proteins. It is necessary to define optimal isolation and culturing conditions for good HSCs yield and proper phenotype preservation for studying the activation of HSCs in vitro. In the present study, the optimal conditions of HSC isolation and culture were examined to maintain the HSC’s undifferentiated phenotype. HSCs were isolated from Balb/c mice liver using Nycodenz, 8, 9.6, and 11%. The efficiency of the isolation procedure was evaluated by cell counting and purity determination by flow cytometry. Quiescent HSCs were cultured in test media supplemented with different combinations of fetal bovine serum (FBS), glutamine (GLN), vitamin A (vitA), insulin, and glucose. The cells were assessed at days 3 and 7 of culture by evaluating the morphology, proliferation using cell counting kit-8, lipid storage using Oil Red O (ORO) staining, expression of a-smooth muscle actin, collagen I, and lecithin-retinol acyltransferase by qRT-PCR and immunocytochemistry (ICC). The results showed that Nycodenz, at 9.6%, yielded the best purity and quantity of HSCs. Maintenance of HSC undifferentiated phenotype was achieved optimizing culturing conditions (serum-free Dulbecco’s Modified Eagle’s Medium (DMEM) supplemented with glucose (100 mg/dl), GLN (0.5 mM), vitA (100 μM), and insulin (50 ng/ml)) with a certain degree of proliferation allowing their perpetuation in culture. In conclusion, we have defined optimal conditions for HSCs isolation and culture.
Collapse
|
16
|
Taguchi K, Yamamoto M. The KEAP1-NRF2 System as a Molecular Target of Cancer Treatment. Cancers (Basel) 2020; 13:cancers13010046. [PMID: 33375248 PMCID: PMC7795874 DOI: 10.3390/cancers13010046] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Nuclear factor erythroid-derived 2-like 2 (encoded by the Nfe2l2 gene; NRF2) is a transcription factor that regulates a variety of cytoprotective genes, including antioxidant enzymes, detoxification enzymes, inflammation-related proteins, drug transporters and metabolic enzymes. NRF2 is regulated by unique molecular mechanisms that stem from Kelch-like ECH-associated protein 1 (KEAP1) in response to oxidative and electrophilic stresses. It has been shown that disturbance or perturbation of the NRF2 activation causes and/or exacerbates many kinds of diseases. On the contrary, aberrant activations of NRF2 also provoke intriguing pathologic features, especially in cancers. Cancer cells with high NRF2 activity have been referred to as NRF2-addicted cancers, which are frequently found in lung cancers. In this review, we summarize the current accomplishments of the KEAP1–NRF2 pathway analyses in special reference to the therapeutic target of cancer therapy. The concept of synthetic lethality provides a new therapeutic approach for NRF2-addicted cancers. Abstract The Kelch-like ECH-associated protein 1 (KEAP1)—Nuclear factor erythroid-derived 2-like 2 (encoded by the Nfe2l2 gene; NRF2) system attracts extensive interest from scientists in basic and clinical cancer research fields, as NRF2 exhibits activity as both an oncogene and tumor suppressor, depending on the context. Especially unique and malignant, NRF2-addicted cancers exhibit high levels of NRF2 expression. Somatic mutations identified in the NRF2 or KEAP1 genes of NRF2-addicted cancers cause the stabilization and accumulation of NRF2. NRF2-addicted cancers hijack the intrinsic roles that NRF2 plays in cytoprotection, including antioxidative and anti-electrophilic responses, as well as metabolic reprogramming, and acquire a marked advantage to survive under severe and limited microenvironments. Therefore, NRF2 inhibitors are expected to have therapeutic effects in patients with NRF2-addicted cancers. In contrast, NRF2 activation in host immune cells exerts significant suppression of cancer cell growth, indicating that NRF2 inducers also have the potential to be therapeutics for cancers. Thus, the KEAP1–NRF2 system makes a broad range of contributions to both cancer development and suppression. These observations thus demonstrate that both NRF2 inhibitors and inducers are useful for the treatment of cancers with high NRF2 activity.
Collapse
Affiliation(s)
- Keiko Taguchi
- Department of Medical Biochemistry, Graduate School of Medicine, Tohoku University, Sendai 980-8575, Japan;
- Department of Medical Biochemistry, Tohoku Medical Megabank Organization, Tohoku University, Sendai 980-8573, Japan
- Advanced Research Center for Innovations in Next-Generation Medicine (INGEM), Tohoku University, Sendai 980-8573, Japan
| | - Masayuki Yamamoto
- Department of Medical Biochemistry, Graduate School of Medicine, Tohoku University, Sendai 980-8575, Japan;
- Department of Medical Biochemistry, Tohoku Medical Megabank Organization, Tohoku University, Sendai 980-8573, Japan
- Advanced Research Center for Innovations in Next-Generation Medicine (INGEM), Tohoku University, Sendai 980-8573, Japan
- Correspondence: ; Tel.: +81-22-728-3039
| |
Collapse
|
17
|
Regulation of basal expression of hepatic PEPCK and G6Pase by AKT2. Biochem J 2020; 477:1021-1031. [PMID: 32096546 DOI: 10.1042/bcj20190570] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 02/04/2020] [Accepted: 02/25/2020] [Indexed: 01/11/2023]
Abstract
Hepatic glucose metabolism signaling downstream of insulin can diverge to multiple pathways including AKT. Genetic studies suggest that AKT is necessary for insulin to suppress gluconeogenesis. To specifically address the role of AKT2, the dominant liver isoform of AKT in the regulation of gluconeogenesis genes, we generated hepatocytes lacking AKT2 (Akt2-/-). We found that, in the absence of insulin signal, AKT2 is required for maintaining the basal level expression of phosphoenolpyruvate carboxyl kinase (PEPCK) and to a lesser extent G6Pase, two key rate-limiting enzymes for gluconeogenesis that support glucose excursion due to pyruvate loading. We further showed that this function of AKT2 is mediated by the phosphorylation of cyclic AMP response element binding (CREB). Phosphorylation of CREB by AKT2 is needed for CREB to induce the expression of PEPCK and likely represents a priming event for unstimulated cells to poise to receive glucagon and other signals. The inhibition of gluconeogenesis by insulin is also dependent on the reduced FOXO1 transcriptional activity at the promoter of PEPCK. When insulin signal is absent, this activity appears to be inhibited by AKT2 in manner that is independent of its phosphorylation by AKT. Together, this action of AKT2 on FOXO1 and CREB to maintain basal gluconeogenesis activity may provide fine-tuning for insulin and glucocorticoid/glucagon to regulate gluconeogenesis in a timely manner to meet metabolic needs.
Collapse
|
18
|
Zhu L, Mou Q, Wang Y, Zhu Z, Cheng M. Resveratrol contributes to the inhibition of liver fibrosis by inducing autophagy via the microRNA‑20a‑mediated activation of the PTEN/PI3K/AKT signaling pathway. Int J Mol Med 2020; 46:2035-2046. [PMID: 33125088 PMCID: PMC7595670 DOI: 10.3892/ijmm.2020.4748] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 09/02/2020] [Indexed: 12/11/2022] Open
Abstract
Liver fibrosis (LF) is a healing response to wounds resulting in liver injury that can cause liver failure or even cancer without functional prevention. Resveratrol (RSV) has been suggested to exert biological effects against various human diseases. MicroRNA-20a (miRNA/miR-20a) has been shown to promote disease progression. The present study aimed to assess the mechanisms through which RSV induces autophagy and activates the miR-20a-mediated phosphatase and tensin homolog (PTEN)/PI3K/AKT signaling pathway in LF. First, a rat model of carbon tetrachlo-ride (CCL4)-induced LF and a cell model of platelet-derived growth factor (PDGF)-BB-stimulated HSC-T6 cells were established for use in subsequent experiments. Subsequently, RSV at a range of concentrations was injected into the model rats with LF. Indicators related to liver injury, oxidative stress and fibrosis were determined in the rats with LF. The RSV-treated HSC-T6 cells were subjected to transfection with miR-20a mimic and PTEN overexpression plasmid to assess the levels of liver injury and LF. A dual-luciferase reporter gene assay was performed to verify the binding sites between PTEN and miR-20a. RSV was found to alleviate LF in rats, and autophagy was enhanced in the rats with LF following RSV treatment. Furthermore, the activation of the PTEN/PI3K/AKT axis attenuated LF, which was reversed by transfection with miR-20a mimic. RSV reversed the inhibitory effects of miR-20a on PTEN expression, reducing miR-20a expression and promoting PTEN, PI3K and p-AKT protein expression, thus attenuating LF. On the whole, the present study demonstrates that RSV induces autophagy and activates the miR-20a-mediated PTEN/PI3K/AKT signaling pathway to attenuate LF. These findings may lead to the development of potential therapeutic strategies for LF.
Collapse
Affiliation(s)
- Lili Zhu
- Department of Blood Transfusion, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Qiuju Mou
- Department of Blood Transfusion, The Affiliated Baiyun Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Yinghui Wang
- Graduate School, Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Zixin Zhu
- Graduate School, Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Mingliang Cheng
- Department of Infectious Diseases, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| |
Collapse
|
19
|
Azar F, Courtet K, Dekky B, Bonnier D, Dameron O, Colige A, Legagneux V, Théret N. Integration of miRNA-regulatory networks in hepatic stellate cells identifies TIMP3 as a key factor in chronic liver disease. Liver Int 2020; 40:2021-2033. [PMID: 32306499 DOI: 10.1111/liv.14476] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 04/08/2020] [Accepted: 04/09/2020] [Indexed: 02/13/2023]
Abstract
BACKGROUND & AIMS Activation of hepatic stellate cells (HSC) is a critical process involved in liver fibrosis. Several miRNAs are implicated in gene regulation during this process but their exact and respective contribution is still incompletely understood. Here we propose an integrative approach of miRNA-regulatory networks to predict new targets. METHODS miRNA regulatory networks in activated HSCs were built using lists of validated miRNAs and the CyTargetLinker tool. The resulting graphs were filtered according to public transcriptomic data and the reduced graphs were analysed through GO annotation. A miRNA network regulating the expression of TIMP3 was further studied in human liver samples, isolated hepatic cells and mouse model of liver fibrosis. RESULTS Within the up-regulated miRNAs, we identified a subnetwork of five miRNAs (miR-21-5p, miR-222-3p, miR-221-3p miR-181b-5p and miR-17-5p) that target TIMP3. We demonstrated that TIMP3 expression is inversely associated with inflammatory activity and IL1-ß expression in vivo. We further showed that IL1-ß inhibits TIMP3 expression in HSC-derived LX-2 cells. Using data from The Cancer Genome Atlas (TCGA), we showed that, in hepatocellular carcinoma (HCC), TIMP3 expression is associated with survival (P < .001), while miR-221 (P < .05), miR-222 (P < .01) and miR-181b (P < .01) are markers for a poor prognosis. CONCLUSIONS Several miRNAs targeting TIMP3 are up-regulated in activated HSCs and down-regulation of TIMP3 expression is associated with inflammatory activity in liver fibrosis and poor prognosis in HCC. The regulatory network including specific miRNAs and TIMP3 is therefore central for the evolution of chronic liver disease.
Collapse
Affiliation(s)
- Fida Azar
- University Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, Rennes, France
| | - Kevin Courtet
- University Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, Rennes, France.,University Rennes, CNRS, IRISA (Institut de recherche en informatique et système aléatoire, Rennes, France
| | - Bassil Dekky
- University Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, Rennes, France
| | - Dominique Bonnier
- University Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, Rennes, France
| | - Olivier Dameron
- University Rennes, CNRS, IRISA (Institut de recherche en informatique et système aléatoire, Rennes, France
| | - Alain Colige
- Laboratory of Connective Tissues Biology, GIGA-R, University of Liege, Sart Tilman, Belgium
| | - Vincent Legagneux
- University Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, Rennes, France
| | - Nathalie Théret
- University Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, Rennes, France.,University Rennes, CNRS, IRISA (Institut de recherche en informatique et système aléatoire, Rennes, France
| |
Collapse
|
20
|
MicroRNA-221: A Fine Tuner and Potential Biomarker of Chronic Liver Injury. Cells 2020; 9:cells9081767. [PMID: 32717951 PMCID: PMC7464779 DOI: 10.3390/cells9081767] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 07/10/2020] [Accepted: 07/10/2020] [Indexed: 12/12/2022] Open
Abstract
The last decade has witnessed significant advancements in our understanding of how small noncoding RNAs, such as microRNAs (miRNAs), regulate disease progression. One such miRNA, miR-221, has been shown to play a key role in the progression of liver fibrosis, a common feature of most liver diseases. Many reports have demonstrated the upregulation of miR-221 in liver fibrosis caused by multiple etiologies such as viral infections and nonalcoholic steatohepatitis. Inhibition of miR-221 via different strategies has shown promising results in terms of the suppression of fibrogenic gene signatures in vitro, as well as in vivo, in independent mouse models of liver fibrosis. In addition, miR-221 has also been suggested as a noninvasive serum biomarker for liver fibrosis and cirrhosis. In this review, we discuss the biology of miR-221, its significance and use as a biomarker during progression of liver fibrosis, and finally, potential and robust approaches that can be utilized to suppress liver fibrosis via inhibition of miR-221.
Collapse
|
21
|
AKT1 Regulates Endoplasmic Reticulum Stress and Mediates the Adaptive Response of Pancreatic β Cells. Mol Cell Biol 2020; 40:MCB.00031-20. [PMID: 32179553 DOI: 10.1128/mcb.00031-20] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 03/12/2020] [Indexed: 12/31/2022] Open
Abstract
Isoforms of protein kinase B (also known as AKT) play important roles in mediating insulin and growth factor signals. Previous studies have suggested that the AKT2 isoform is critical for insulin-regulated glucose metabolism, while the role of the AKT1 isoform remains less clear. This study focuses on the effects of AKT1 on the adaptive response of pancreatic β cells. Using a mouse model with inducible β-cell-specific deletion of the Akt1 gene (βA1KO mice), we showed that AKT1 is involved in high-fat-diet (HFD)-induced growth and survival of β cells but is unnecessary for them to maintain a population in the absence of metabolic stress. When unchallenged, βA1KO mice presented the same metabolic profile and β-cell phenotype as the control mice with an intact Akt1 gene. When metabolic stress was induced by HFD, β cells in control mice with intact Akt1 proliferated as a compensatory mechanism for metabolic overload. Similar effects were not observed in βA1KO mice. We further demonstrated that AKT1 protein deficiency caused endoplasmic reticulum (ER) stress and potentiated β cells to undergo apoptosis. Our results revealed that AKT1 protein loss led to the induction of eukaryotic initiation factor 2 α subunit (eIF2α) signaling and ER stress markers under normal-chow-fed conditions, indicating chronic low-level ER stress. Together, these data established a role for AKT1 as a growth and survival factor for adaptive β-cell response and suggest that ER stress induction is responsible for this effect of AKT1.
Collapse
|
22
|
Wen Y, Chen J, Li J, Arif W, Kalsotra A, Irudayaraj J. Effect of PFOA on DNA Methylation and Alternative Splicing in Mouse Liver. Toxicol Lett 2020; 329:38-46. [PMID: 32320774 DOI: 10.1016/j.toxlet.2020.04.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 03/13/2020] [Accepted: 04/16/2020] [Indexed: 02/07/2023]
Abstract
Perfluorooctanoic acid (PFOA) is a persistent organic pollutant prevalent in the environment and implicated in damage to the liver leading to a fatty liver phenotype called hepatocellular steatosis. Our goal is to provide a basis for PFOA-induced hepatocellular steatosis in relation to epigenetic alterations and mRNA splicing. Young adult female mice exposed to different concentrations of PFOA showed an increase in liver weight with decreased global DNA methylation (5-mC). At higher concentrations, the expression of DNA methyltransferase 3A (Dnmt3a) was significantly reduced and the expression of tet methycytosine dioxygenase 1 (Tet1) was significantly increased. There was no significant change in the other Dnmts and Tets. PFOA exposure significantly increased the expression of cell cycle regulators and anti-apoptotic genes. The expression of multiple genes involved in mTOR (mammalian target of rapamycin) signaling pathway were altered significantly with reduction in Pten (phosphatase and tensin homolog, primary inhibitor of mTOR pathway) expression. Multiple splicing factors whose protein but not mRNA levels affected by PFOA exposure were identified. The changes in protein abundance of the splicing factors was also reflected in altered splicing pattern of their target genes, which provided new insights on the previously unexplored mechanisms of PFOA-mediated hepatotoxicity and pathogenesis.
Collapse
Affiliation(s)
- Yi Wen
- Department of Bioengineering. University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA; Biomedical Research Center in Mills Breast Cancer Institute, Carle Foundation Hospital, Urbana, IL 61801, USA; Cancer Center at Illinois (CCIL), University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Jackie Chen
- Department of Biochemistry, School of Molecular and Cell Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Junya Li
- Department of Biochemistry, School of Molecular and Cell Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Waqar Arif
- Department of Biochemistry, School of Molecular and Cell Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Auinash Kalsotra
- Department of Biochemistry, School of Molecular and Cell Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Cancer Center at Illinois (CCIL), University of Illinois at Urbana-Champaign, Urbana, IL, USA; Carl R. Woese Institute of Genomic Biology, University of Illinois, Urbana-Champaign, Illinois, USA
| | - Joseph Irudayaraj
- Department of Bioengineering. University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA; Biomedical Research Center in Mills Breast Cancer Institute, Carle Foundation Hospital, Urbana, IL 61801, USA; Micro and Nanotechnology Laboratory. University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA; Cancer Center at Illinois (CCIL), University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
23
|
Liang H, Wang X, Si C, Duan Y, Chen B, Liang H, Yang D. Downregulation of miR‑141 deactivates hepatic stellate cells by targeting the PTEN/AKT/mTOR pathway. Int J Mol Med 2020; 46:406-414. [PMID: 32319536 DOI: 10.3892/ijmm.2020.4578] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 03/27/2020] [Indexed: 01/18/2023] Open
Abstract
The activation of hepatic stellate cells (HSCs) caused by stimulating factors or fibrogenic cytokines is the critical stage of liver fibrosis. Recent studies have demonstrated the influence of microRNAs (miRNAs or miRs) on HSC activation and transformation; however, the function and underlying mechanisms of miRNAs in HSC activation have not yet been completely clarified. In the present study, transforming growth factor β1 (TGF‑β1) was used to treat human HSC lines (HSC‑T6 and LX2 cells) to simulate the activation of HSCs in vivo and whether the expression of miRNAs in HSCs was affected by TGF‑β1 treatment was examined using a miRNA microarray. It was observed that miR‑141 was one of the most upregulated miRNAs during HSC activation. Functional analyses revealed that miR‑141 knockdown suppressed the viability of HSCs and inhibited the expression levels of pro‑fibrotic markers. In addition, phosphatase and tensin homolog (PTEN), a well‑known suppressor of the AKT/mammalian target of rapamycin (mTOR) pathway, was found to be directly targeted by miR‑141 in HSCs. More importantly, the knockdown of PTEN markedly reversed the suppressive effects of miR‑141 inhibition on the viability of and the expression levels of pro‑fibrotic markers during HSC activation. Finally, it was observed that the downregulation of miR‑141 blocked the TGF‑β1‑induced activation of the AKT/mTOR pathway in HSCs. On the whole, the findings of the present study indicate that miR‑141 inhibition suppresses HSC activation via the AKT/mTOR pathway by targeting PTEN, highlighting that miR‑141 may serve as a novel therapeutic target for liver fibrosis.
Collapse
Affiliation(s)
- Haijun Liang
- Department of Infectious Disease, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
| | - Xinwei Wang
- Department of Infectious Disease, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
| | - Changyun Si
- Department of Infectious Disease, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
| | - Yuxiu Duan
- Department of Infectious Disease, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
| | - Baoxin Chen
- Department of Infectious Disease, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
| | - Haixia Liang
- Department of Neurology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
| | - Daokun Yang
- Department of Infectious Disease, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
| |
Collapse
|
24
|
Tu T, Chen J, Chen L, Stiles BL. Dual-Specific Protein and Lipid Phosphatase PTEN and Its Biological Functions. Cold Spring Harb Perspect Med 2020; 10:cshperspect.a036301. [PMID: 31548229 DOI: 10.1101/cshperspect.a036301] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Phosphatase and tensin homolog deleted on chromosome 10 (PTEN) encodes a 403-amino acid protein with an amino-terminal domain that shares sequence homology with the actin-binding protein tensin and the putative tyrosine-protein phosphatase auxilin. Crystal structure analysis of PTEN has revealed a C2 domain that binds to phospholipids in membranes and a phosphatase domain that displays dual-specific activity toward both tyrosine (Y), serine (S)/threonine (T), as well as lipid substrates in vitro. Characterized primarily as a lipid phosphatase, PTEN plays important roles in multiple cellular processes including cell growth/survival as well as metabolism.
Collapse
Affiliation(s)
- Taojian Tu
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California 90033, USA
| | - Jingyu Chen
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California 90033, USA
| | - Lulu Chen
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California 90033, USA
| | - Bangyan L Stiles
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California 90033, USA.,Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, California 90033, USA
| |
Collapse
|
25
|
Walter LA, Lin YH, Halbrook CJ, Chuh KN, He L, Pedowitz NJ, Batt AR, Brennan CK, Stiles BL, Lyssiotis CA, Pratt MR. Inhibiting the Hexosamine Biosynthetic Pathway Lowers O-GlcNAcylation Levels and Sensitizes Cancer to Environmental Stress. Biochemistry 2019; 59:3169-3179. [PMID: 31625393 DOI: 10.1021/acs.biochem.9b00560] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The amounts of the intracellular glycosylation, O-GlcNAc modification, are increased in essentially all tumors when compared to healthy tissue, and lowering O-GlcNAcylation levels results in reduced tumorigenesis and increased cancer cell death. Therefore, the pharmacological reduction of O-GlcNAc may represent a therapeutic vulnerability. The most direct approach to this goal is the inhibition of O-GlcNAc transferase (OGT), the enzyme that directly adds the modification to proteins. However, despite some recent success, this enzyme has proven difficult to inhibit. An alternative strategy involves starving OGT of its sugar substrate UDP-GlcNAc by targeting enzymes of the hexosamine biosynthetic pathway (HBP). Here, we explore the potential of the rate-determining enzyme of this pathway, glutamine fructose-6-phosphate amidotransferase (GFAT). We first show that CRISPR-mediated knockout of GFAT results in inhibition of cancer cell growth in vitro and a xenograft model that correlates with O-GlcNAcylation levels. We then demonstrate that pharmacological inhibition of GFAT sensitizes a small panel of cancer cells to undergo apoptosis in response to diamide-induced oxidative stress. Finally, we find that GFAT expression and O-GlcNAc levels are increased in a spontaneous mouse model of liver cancer. Together these experiments support the further development of inhibitors of the HBP as an indirect approach to lowering O-GlcNAcylation levels in cancer.
Collapse
|
26
|
Wu JC, Chen R, Luo X, Li ZH, Luo SZ, Xu MY. MicroRNA-194 inactivates hepatic stellate cells and alleviates liver fibrosis by inhibiting AKT2. World J Gastroenterol 2019; 25:4468-4480. [PMID: 31496625 PMCID: PMC6710173 DOI: 10.3748/wjg.v25.i31.4468] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 06/25/2019] [Accepted: 07/19/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Activation of hepatic stellate cells (HSCs) is a pivotal event in the onset and progression of liver fibrosis. Loss of microRNA-194 (miR-194) has been reported in activated HSCs, but the actual role of miR-194 in liver fibrosis remains uncertain.
AIM To explore the role and potential mechanism of miR-194-mediated regulation of liver fibrosis in vitro and in vivo.
METHODS The expression of miR-194 was examined in human fibrotic liver tissues, activated HSCs, and a carbon tetrachloride (CCl4) mouse model by qPCR. The effects of AKT2 regulation by miR-194 on the activation and proliferation of HSCs were assessed in vitro. For in vivo experiments, we reintroduced miR-194 in mice using a miR-194 agomir to investigate the functions of miR-194 in liver fibrosis.
RESULTS MiR-194 expression was notably lacking in activated HSCs from both humans and mice. Overexpression of miR-194 (OV-miR-194) inhibited α-smooth muscle actin (α-SMA) and type I collagen (Col I) expression and suppressed cell proliferation in HSCs by causing cell cycle arrest in G0/G1 phase. AKT2 was predicted to be a target of miR-194. Notably, the effects of miR-194 knockdown in HSCs were almost blocked by AKT2 deletion, indicating that miR-194 plays a role in HSCs via regulation of AKT2. Finally, miR-194 agomir treatment dramatically ameliorated liver fibrosis in CCl4-treated mice.
CONCLUSION We revealed that miR-194 plays a protective role by inhibiting the activation and proliferation of HSCs via AKT2 suppression. Our results further propose miR-194 as a potential therapeutic target for liver fibrosis.
Collapse
Affiliation(s)
- Jun-Cheng Wu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Rong Chen
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Xin Luo
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Zheng-Hong Li
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Sheng-Zheng Luo
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Ming-Yi Xu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| |
Collapse
|
27
|
Lukaszewicz AI, Nguyen C, Melendez E, Lin DP, Teo JL, Lai KKY, Huttner WB, Shi SH, Kahn M. The Mode of Stem Cell Division Is Dependent on the Differential Interaction of β-Catenin with the Kat3 Coactivators CBP or p300. Cancers (Basel) 2019; 11:cancers11070962. [PMID: 31324005 PMCID: PMC6678591 DOI: 10.3390/cancers11070962] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 07/02/2019] [Accepted: 07/04/2019] [Indexed: 01/03/2023] Open
Abstract
Normal long-term repopulating somatic stem cells (SSCs) preferentially divide asymmetrically, with one daughter cell remaining in the niche and the other going on to be a transient amplifying cell required for generating new tissue in homeostatic maintenance and repair processes, whereas cancer stem cells (CSCs) favor symmetric divisions. We have previously proposed that differential β-catenin modulation of transcriptional activity via selective interaction with either the Kat3 coactivator CBP or its closely related paralog p300, regulates symmetric versus asymmetric division in SSCs and CSCs. We have previously demonstrated that SSCs that divide asymmetrically per force retain one of the dividing daughter cells in the stem cell niche, even when treated with specific CBP/β-catenin antagonists, whereas CSCs can be removed from their niche via forced stochastic symmetric differentiative divisions. We now demonstrate that loss of p73 in early corticogenesis biases β-catenin Kat3 coactivator usage and enhances β-catenin/CBP transcription at the expense of β-catenin/p300 transcription. Biased β-catenin coactivator usage has dramatic consequences on the mode of division of neural stem cells (NSCs), but not neurogenic progenitors. The observed increase in symmetric divisions due to enhanced β-catenin/CBP interaction and transcription leads to an immediate increase in NSC symmetric differentiative divisions. Moreover, we demonstrate for the first time that the complex phenotype caused by the loss of p73 can be rescued in utero by treatment with the small-molecule-specific CBP/β-catenin antagonist ICG-001. Taken together, our results demonstrate the causal relationship between the choice of β-catenin Kat3 coactivator and the mode of stem cell division.
Collapse
Affiliation(s)
- Agnes I Lukaszewicz
- Department of Biochemistry and Molecular Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Cu Nguyen
- Center for Molecular Pathways and Drug Discovery, University of Southern California, Los Angeles, CA 90033, USA
- Department of Molecular Medicine, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Elizabeth Melendez
- Center for Molecular Pathways and Drug Discovery, University of Southern California, Los Angeles, CA 90033, USA
- Department of Molecular Medicine, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - David P Lin
- Department of Molecular Medicine, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Jia-Ling Teo
- Center for Molecular Pathways and Drug Discovery, University of Southern California, Los Angeles, CA 90033, USA
- Department of Molecular Medicine, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Keane K Y Lai
- Department of Molecular Medicine, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
- Department of Pathology, City of Hope National Medical Center, Duarte, CA 91010, USA
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Wieland B Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden 01307, Germany
| | - Song-Hai Shi
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Michael Kahn
- Department of Biochemistry and Molecular Medicine, University of Southern California, Los Angeles, CA 90033, USA.
- Center for Molecular Pathways and Drug Discovery, University of Southern California, Los Angeles, CA 90033, USA.
- Department of Molecular Medicine, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA.
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA.
- Department of Molecular Pharmacology and Toxicology, University of Southern California, Los Angeles, CA 90033, USA.
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA.
| |
Collapse
|
28
|
Gonçalves LR, de Oliveira da Silva B, Scarinci LD, Ramos LF, Moraes KC. MicroRNA-1254 contributes to the controlling of pro-fibrogenic environment in LX-2 cells by modulating SMAD3 and wound repair: new insights in hepatic fibrosis. Cell Biol Int 2019; 43:333-343. [DOI: 10.1002/cbin.11075] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 11/15/2018] [Indexed: 12/15/2022]
Affiliation(s)
- Letícia Rocha Gonçalves
- Laboratório de Biologia Molecular, Departamento de Biologia; Instituto de Biociências, Universidade Estadual Paulista “Júlio de Mesquita Filho” − Campus Rio Claro; Rio Claro SP 13506-900 Brazil
| | | | - Letícia Destefani Scarinci
- Laboratório de Biologia Molecular, Departamento de Biologia; Instituto de Biociências, Universidade Estadual Paulista “Júlio de Mesquita Filho” − Campus Rio Claro; Rio Claro SP 13506-900 Brazil
| | - Letícia Ferreira Ramos
- Laboratório de Biologia Molecular, Departamento de Biologia; Instituto de Biociências, Universidade Estadual Paulista “Júlio de Mesquita Filho” − Campus Rio Claro; Rio Claro SP 13506-900 Brazil
| | - Karen C.M. Moraes
- Laboratório de Biologia Molecular, Departamento de Biologia; Instituto de Biociências, Universidade Estadual Paulista “Júlio de Mesquita Filho” − Campus Rio Claro; Rio Claro SP 13506-900 Brazil
| |
Collapse
|
29
|
Kumar P, Raeman R, Chopyk DM, Smith T, Verma K, Liu Y, Anania FA. Adiponectin inhibits hepatic stellate cell activation by targeting the PTEN/AKT pathway. Biochim Biophys Acta Mol Basis Dis 2018; 1864:3537-3545. [PMID: 30293572 PMCID: PMC6529190 DOI: 10.1016/j.bbadis.2018.08.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 08/05/2018] [Accepted: 08/07/2018] [Indexed: 02/08/2023]
Abstract
Adiponectin inhibits hepatic stellate cell (HSC) activation and subsequent development of liver fibrosis via multiple mechanisms. Phosphatase and tensin homolog deletion 10 (PTEN) plays a crucial role in suppression of HSC activation, but its regulation by adiponectin is not fully understood. Here, we investigated the effect of adiponectin on PTEN in LX-2 cells, a human cell line and examined the underlying molecular mechanisms involved in adiponectin-mediated upregulation of PTEN activity during fibrosis. PTEN expression was found to be significantly reduced in the livers of mice treated with CCl4, whereas its expression was rescued by adiponectin treatment. The DNA methylation proteins DNMT1, DNMT3A, and DNMT3B are all highly expressed in activated primary HSCs compared to quiescent HSCs, and thus represent additional regulatory targets during liver fibrogenesis. Expression of DNMT proteins was significantly induced in the presence of fibrotic stimuli; however, only DNMT3B expression was reduced in the presence of adiponectin. Adiponectin-induced suppression of DNMT3B was found to be mediated by enhanced miR-29b expression. Furthermore, PTEN expression was significantly increased by overexpression of miR-29b, whereas its expression was markedly reduced by a miR-29b inhibitor in LX-2 cells. These findings suggest that adiponectin-induced upregulation of miR-29b can suppress DNMT3B transcription in LX-2 cells, thus resulting in reduced methylation of PTEN CpG islands and ultimately suppressing the PI3K/AKT pathway. Together, these data suggest a possible new explanation for the inhibitory effect of adiponectin on HSC activation and liver fibrogenesis.
Collapse
Affiliation(s)
- Pradeep Kumar
- Division of Digestive Diseases, Department of Medicine, Emory University, Atlanta, GA, USA.
| | - Reben Raeman
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Daniel M Chopyk
- Division of Digestive Diseases, Department of Medicine, Emory University, Atlanta, GA, USA
| | - Tekla Smith
- Division of Digestive Diseases, Department of Medicine, Emory University, Atlanta, GA, USA
| | - Kiran Verma
- Labratory of Biochemical Pharmacology, Department of Pediatrics, Emory University, Atlanta, GA, USA
| | - Yunshan Liu
- Division of Digestive Diseases, Department of Medicine, Emory University, Atlanta, GA, USA
| | - Frank A Anania
- Division of Digestive Diseases, Department of Medicine, Emory University, Atlanta, GA, USA
| |
Collapse
|
30
|
Elevated Nrf-2 responses are insufficient to mitigate protein carbonylation in hepatospecific PTEN deletion mice. PLoS One 2018; 13:e0198139. [PMID: 29799837 PMCID: PMC5969769 DOI: 10.1371/journal.pone.0198139] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 05/14/2018] [Indexed: 12/20/2022] Open
Abstract
Objective In the liver, a contributing factor in the pathogenesis of non-alcoholic fatty liver disease (NASH) is oxidative stress, which leads to the accumulation of highly reactive electrophilic α/β unsaturated aldehydes. The objective of this study was to determine the impact of NASH on protein carbonylation and antioxidant responses in a murine model. Methods Liver-specific phosphatase and tensin homolog (PTEN)-deletion mice (PTENLKO) or control littermates were fed a standard chow diet for 45–55 weeks followed by analysis for liver injury, oxidative stress and inflammation. Results Histology and Picrosirius red-staining of collagen deposition within the extracellular matrix revealed extensive steatosis and fibrosis in the PTENLKO mice but no steatosis or fibrosis in controls. Increased steatosis and fibrosis corresponded with significant increases in inflammation. PTEN-deficient livers showed significantly increased cell-specific oxidative damage, as detected by 4-hydroxy-2-nonenal (4-HNE) and acrolein staining. Elevated staining correlated with an increase in nuclear DNA repair foci (γH2A.X) and cellular proliferation index (Ki67) within zones 1 and 3, indicating oxidative damage was zonally restricted and was associated with increased DNA damage and cell proliferation. Immunoblots showed that total levels of antioxidant response proteins induced by nuclear factor erythroid-2-like-2 (Nrf2), including GSTμ, GSTπ and CBR1/3, but not HO-1, were elevated in PTENLKO as compared to controls, and IHC showed this response also occurred only in zones 1 and 3. Furthermore, an analysis of autophagy markers revealed significant elevation of p62 and LC3II expression. Mass spectrometric (MS) analysis identified significantly more carbonylated proteins in whole cell extracts prepared from PTENLKO mice (966) as compared to controls (809). Pathway analyses of identified proteins did not uncover specific pathways that were preferentially carbonylated in PTENLKO livers but, did reveal specific strongly increased carbonylation of thioredoxin reductase and of glutathione-S-transferases (GST) M6, O1, and O2. Conclusions Results show that disruption of PTEN resulted in steatohepatitis, fibrosis and caused hepatic induction of the Nrf2-dependent antioxidant system at least in part due to elevation of p62. This response was both cell-type and zone specific. However, these responses were insufficient to mitigate the accumulation of products of lipid peroxidation.
Collapse
|
31
|
Li NN, Meng XS, Men WX, Bao YR, Wang S. Total Flavonoids from Oroxylum indicum Induce Apoptosis via PI3K/Akt/PTEN Signaling Pathway in Liver Cancer. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2018; 2018:3021476. [PMID: 29636773 PMCID: PMC5832136 DOI: 10.1155/2018/3021476] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 01/12/2018] [Accepted: 01/16/2018] [Indexed: 12/13/2022]
Abstract
Total flavonoids (TF), derived from the seeds of Oroxylum indicum (L.) Vent., possess many pharmacological functions. In the present study, H22-bearing mice and SMMC-7721 models were employed to evaluate the antitumor activity of TF and to and investigate its possible mechanisms both in vitro and in vivo. Cell viability was evaluated by MTT assay; cell apoptosis rate was analyzed via Annexin V-FITC/PI double staining by flow cytometer. Meanwhile, the expressions of apoptosis-related mRNA and proteins were evaluated by RT-PCR and Western blot analysis. The results revealed that TF could significantly inhibit the tumor growth, and the possible mechanism was related to the effect of inducing tumor cells apoptosis through PI3K/Akt/PTEN signaling pathway. This study has provided a theoretical basis for the further development and application of TF as antitumor drugs.
Collapse
Affiliation(s)
- Nan-nan Li
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China
| | - Xian-sheng Meng
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China
- Component Medicine Engineering Research Center of Liaoning Province, Dalian 116600, China
- Liaoning Province Modern Chinese Medicine Research Engineering Laboratory, Dalian 116600, China
| | - Wen-xiao Men
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China
| | - Yong-rui Bao
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China
- Component Medicine Engineering Research Center of Liaoning Province, Dalian 116600, China
- Liaoning Province Modern Chinese Medicine Research Engineering Laboratory, Dalian 116600, China
| | - Shuai Wang
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China
- Component Medicine Engineering Research Center of Liaoning Province, Dalian 116600, China
- Liaoning Province Modern Chinese Medicine Research Engineering Laboratory, Dalian 116600, China
| |
Collapse
|
32
|
Chen CY, Chen J, He L, Stiles BL. PTEN: Tumor Suppressor and Metabolic Regulator. Front Endocrinol (Lausanne) 2018; 9:338. [PMID: 30038596 PMCID: PMC6046409 DOI: 10.3389/fendo.2018.00338] [Citation(s) in RCA: 376] [Impact Index Per Article: 53.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 06/05/2018] [Indexed: 12/19/2022] Open
Abstract
Phosphatase and Tensin Homolog deleted on Chromosome 10 (PTEN) is a dual phosphatase with both protein and lipid phosphatase activities. PTEN was first discovered as a tumor suppressor with growth and survival regulatory functions. In recent years, the function of PTEN as a metabolic regulator has attracted significant attention. As the lipid phosphatase that dephosphorylates phosphatidylinositol-3, 4, 5-phosphate (PIP3), PTEN reduces the level of PIP3, a critical 2nd messenger mediating the signal of not only growth factors but also insulin. In this review, we introduced the discovery of PTEN, the PTEN-regulated canonical and nuclear signals, and PTEN regulation. We then focused on the role of PTEN and PTEN-regulated signals in metabolic regulation. This included the role of PTEN in glycolysis, gluconeogenesis, glycogen synthesis, lipid metabolism as well as mitochondrial metabolism. We also included how PTEN and PTEN regulated metabolic functions may act paradoxically toward insulin sensitivity and tumor metabolism and growth. Further understanding of how PTEN regulates metabolism and how such regulations lead to different biological outcomes is necessary for interventions targeting at the PTEN-regulated signals in either cancer or diabetes treatment.
Collapse
Affiliation(s)
- Chien-Yu Chen
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, United States
| | - Jingyu Chen
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, United States
| | - Lina He
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, United States
| | - Bangyan L. Stiles
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, United States
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- *Correspondence: Bangyan L. Stiles
| |
Collapse
|
33
|
Luo X, Liao R, Hanley KL, Zhu HH, Malo KN, Hernandez C, Wei X, Varki NM, Alderson N, Chu C, Li S, Fan J, Loomba R, Qiu SJ, Feng GS. Dual Shp2 and Pten Deficiencies Promote Non-alcoholic Steatohepatitis and Genesis of Liver Tumor-Initiating Cells. Cell Rep 2017; 17:2979-2993. [PMID: 27974211 DOI: 10.1016/j.celrep.2016.11.048] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 10/20/2016] [Accepted: 11/15/2016] [Indexed: 01/14/2023] Open
Abstract
The complexity of liver tumorigenesis is underscored by the recently observed anti-oncogenic effects of oncoproteins, although the mechanisms are unclear. Shp2/Ptpn11 is a proto-oncogene in hematopoietic cells and antagonizes the effect of tumor suppressor Pten in leukemogenesis. In contrast, we show here cooperative functions of Shp2 and Pten in suppressing hepatocarcinogenesis. Ablating both Shp2 and Pten in hepatocytes induced early-onset non-alcoholic steatohepatitis (NASH) and promoted genesis of liver tumor-initiating cells likely due to augmented cJun expression/activation and elevated ROS and inflammation in the hepatic microenvironment. Inhibiting cJun partially suppressed NASH-driven liver tumorigenesis without improving NASH. SHP2 and PTEN deficiencies were detected in liver cancer patients with poor prognosis. These data depict a mechanism of hepato-oncogenesis and suggest a potential therapeutic strategy.
Collapse
Affiliation(s)
- Xiaolin Luo
- Department of Pathology, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Rui Liao
- Department of Pathology, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Department of Hepatobiliary Surgery, The First Affiliated Hospital, Chongqing Medical University, Chongqing 40016, China
| | - Kaisa L Hanley
- Molecular Biology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Helen He Zhu
- Department of Pathology, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Kirsten N Malo
- Molecular Biology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Carolyn Hernandez
- Department of Medicine, NAFLD Research Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Xufu Wei
- Department of Pathology, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Department of Hepatobiliary Surgery, The First Affiliated Hospital, Chongqing Medical University, Chongqing 40016, China
| | - Nissi M Varki
- Department of Pathology, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Nazilla Alderson
- Department of Pathology, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Catherine Chu
- Molecular Biology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Shuangwei Li
- Department of Pathology, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jia Fan
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Rohit Loomba
- Department of Medicine, NAFLD Research Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Shuang-Jian Qiu
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Gen-Sheng Feng
- Department of Pathology, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Molecular Biology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
34
|
Li Y, Xiong L, Gong J. Lyn kinase enhanced hepatic fibrosis by modulating the activation of hepatic stellate cells. Am J Transl Res 2017; 9:2865-2877. [PMID: 28670375 PMCID: PMC5489887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 04/25/2017] [Indexed: 06/07/2023]
Abstract
The non-selectivity of tyrosine kinase inhibitors is the leading cause of drug withdrawals, and limits their application in anti-fibrosis. The role of Src tyrosine kinase Lyn in hepatic fibrosis remains elusive. In this study, we aimed to elucidate the role of Lyn kinase in the pathogenesis of hepatic fibrosis. Through examining Lyn-transgenic (Lyn TG) mice treated with CCl4 (carbon tetrachloride), we determined whether Lyn kinase is involved in the pathogenesis of hepatic fibrosis. On top of that, we also investigated the role of Lyn in the activation of hepatic stellate cells (HSCs) in vitro. Here, we showed that Lyn kinase was highly expressed in liver fibrosis upon CCl4 treatment. Meanwhile, Lyn TG mice showed that perivascular infiltration of mononuclear cells, and the markers of liver injury and hepatocytes apoptosis were significantly increased in liver tissue after CCl4 treatment. In comparison with wild-type (WT) mice after CCl4 treatment, we found that the fibrotic score in liver tissues of Lyn TG mice with the same treatment went up dramatically, so did the gene expression of fibrotic markers. In addition, over-expression of Lyn kinase drastically promoted the expression of HSCs activation markers in vivo or in vitro. Additionally, the Src-specific inhibitor PP2 significantly suppressed the increased expression of integrin αvβ3 in TGF-β1-induced HSCs, and PP2 further induced HSC apoptosis in TGF-β1-treated cells. These results collectively indicated that Lyn kinase is implicated in the pathogenesis of hepatic fibrosis through the modulating of HSC activation.
Collapse
Affiliation(s)
- Yin Li
- The First Clinic College, Chongqing Medical UniversityChongqing 401331, China
- Central Laboratory, The Second Affiliated Hospital of Chongqing Medical UniversityChongqing 400010, China
| | - Lin Xiong
- The School of Laboratory Medicine, Chongqing Medical UniversityChongqing 400016, China
| | - Jianping Gong
- Department of Hepatobiliary Surgery, Second Affiliated Hospital of Chongqing Medical UniversityChongqing 400010, China
| |
Collapse
|
35
|
|
36
|
Jia C, Medina V, Liu C, He L, Qian D, Taojian T, Okamoto CT, Stiles BL. Crosstalk of LKB1- and PTEN-regulated signals in liver morphogenesis and tumor development. Hepatol Commun 2017; 1:153-167. [PMID: 29152604 PMCID: PMC5687583 DOI: 10.1002/hep4.1027] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Liver kinase B 1 (LKB1 or STK11) and phosphatase and tensin homologue deleted on chromosome 10 (PTEN) are two tumor suppressors that regulate the mammalian target of rapamycin signaling pathway. Deletion studies show that loss of either Lkb1 (Lkb+/–) or Pten (PtenloxP/loxP; Alb‐Cre+) leads to liver injury and development of hepatocarcinoma. In this study, we investigated the crosstalk of LKB1 and PTEN loss during tumorigenesis and liver development. We show that haplo‐insufficiency of Lkb1 in the liver leads to advanced tumor development in Pten‐null mice (PtenloxP/loxP; LkbloxP/+; Alb‐Cre+). Our analysis shows that LKB1 and PTEN interact with each other in their regulation of fatty acid synthase as well as p21 expression. The combined loss of LKB1 and PTEN (PtenloxP/loxP; LkbloxP/loxP; Alb‐Cre+) also leads to the inability to form zonal structures in the liver. The lack of metabolic zonal structures is consistent with the inability of the livers to store glycogen as well as elevated plasma bilirubin and alanine aminotransferase, indicative of liver dysfunction. These structural and functional defects are associated with cytoplasm distribution of a canalicular membrane protein multidrug resistant protein 2, which is responsible for clearing bilirubin. This observed regulation of multidrug resistant protein 2 by LKB1 likely contributes to the lack of cellular polarity and the early lethality phenotype associated with the homozygous loss of Lkb1 alone or in combination with Pten. Finally, Pten deletion does not rescue the precocious ductal plate formation reported for Lkb1‐deleted livers. Conclusion: Our study dissected the functional and molecular crosstalk of PTEN and LKB1 and elucidated key molecular targets for such interactions. (Hepatology Communications 2017;1:153‐167)
Collapse
Affiliation(s)
- Chengyou Jia
- Department of Nuclear Medicine, Central Laboratory for Medical Research, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai 200072, China.,Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90033
| | - Vivian Medina
- Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90033
| | - Chenchang Liu
- Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90033
| | - Lina He
- Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90033
| | - Daohai Qian
- Department of Nuclear Medicine, Central Laboratory for Medical Research, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai 200072, China.,Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90033
| | - Tu Taojian
- Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90033
| | - Curtis T Okamoto
- Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90033
| | - Bangyan L Stiles
- Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90033.,Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
| |
Collapse
|
37
|
Activation of Insulin-PI3K/Akt-p70S6K Pathway in Hepatic Stellate Cells Contributes to Fibrosis in Nonalcoholic Steatohepatitis. Dig Dis Sci 2017; 62:968-978. [PMID: 28194671 DOI: 10.1007/s10620-017-4470-9] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 01/20/2017] [Indexed: 01/18/2023]
Abstract
BACKGROUND AND AIMS Hyperinsulinemia and insulin resistance are hallmark features of nonalcoholic fatty liver disease and steatohepatitis (NASH). It remains unclear whether and how insulin contributes to the development of fibrosis in NASH. In this study, we explored insulin signaling in the regulation of hepatic stellate cell (HSC) activation and the progression of NASH-fibrosis. METHODS Phosphorylation of Akt and p70S6K were examined in primary HSC and in a rat model of NASH-fibrosis induced by high-fat and high-cholesterol diet for 24 weeks. HSC activation was analyzed for the changes in cell morphology, intracellular lipid droplets, expression of α-SMA and cell proliferation. The serum markers and histology for NASH-fibrosis were also characterized in animals. RESULTS Insulin enhanced the expression of smooth muscle actin-α in quiescent but not in activated HSC in culture. Insulin-mediated activation of the PI3K/Akt-p70S6K pathway was involved in the regulation of profibrogenic effects of insulin. Although insulin did not stimulate HSC proliferation directly, the insulin-PI3K/Akt-p70S6K pathway was necessary for serum-enhanced cell proliferation during initial HSC activation. In a rat model of NASH-fibrosis induced by high-fat and high-cholesterol diet, hyperinsulinemia is associated with the activation of p70S6K and enhanced fibrosis. CONCLUSION The insulin-PI3K/Akt-p70S6K pathway plays an important role in the early activation of HSC. The profibrogenic effect of insulin is dependent on the activation stage of HSC. Dysregulation of the insulin pathway likely correlates with the development of fibrosis in NASH, suggesting a potentially novel antifibrotic target of inhibiting insulin signaling in HSC.
Collapse
|
38
|
MicroRNA-29a Alleviates Bile Duct Ligation Exacerbation of Hepatic Fibrosis in Mice through Epigenetic Control of Methyltransferases. Int J Mol Sci 2017; 18:ijms18010192. [PMID: 28106784 PMCID: PMC5297823 DOI: 10.3390/ijms18010192] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 01/10/2017] [Accepted: 01/13/2017] [Indexed: 12/17/2022] Open
Abstract
MicroRNA-29 (miR-29) is found to modulate hepatic stellate cells’ (HSCs) activation and, thereby, reduces liver fibrosis pathogenesis. Histone methyltransferase regulation of epigenetic reactions reportedly participates in hepatic fibrosis. This study is undertaken to investigate the miR-29a regulation of the methyltransferase signaling and epigenetic program in hepatic fibrosis progression. miR-29a transgenic mice (miR-29aTg mice) and wild-type littermates were subjected to bile duct-ligation (BDL) to develop cholestatic liver fibrosis. Primary HSCs were transfected with a miR-29a mimic and antisense inhibitor. Profibrogenic gene expression, histone methyltransferases and global genetic methylation were probed with real-time quantitative RT-PCR, immunohistochemical stain, Western blot and ELISA. Hepatic tissue in miR-29aTg mice displayed weak fibrotic matrix as evidenced by Sirius Red staining concomitant with low fibrotic matrix collagen 1α1 expression within affected tissues compared to the wild-type mice. miR-29a overexpression reduced the BDL exaggeration of methyltransferases, DNMT1, DNMT3b and SET domain containing 1A (SET1A) expression. It also elevated phosphatase and tensin homolog deleted on chromosome 10 (PTEN) signaling within liver tissue. In vitro, miR-29a mimic transfection lowered collagen 1α1, DNMT1, DNMT3b and SET1A expression in HSCs. Gain of miR-29a signaling resulted in DNA hypomethylation and high PTEN expression. This study shines a new light on miR-29a inhibition of methyltransferase, a protective effect to maintain the DNA hypomethylation state that decreases fibrogenic activities in HSC. These robust analyses also highlight the miR-29a regulation of epigenetic actions to ameliorate excessive fibrosis during cholestatic liver fibrosis development.
Collapse
|
39
|
Zhang Z, Zhao S, Yao Z, Wang L, Shao J, Chen A, Zhang F, Zheng S. Autophagy regulates turnover of lipid droplets via ROS-dependent Rab25 activation in hepatic stellate cell. Redox Biol 2016; 11:322-334. [PMID: 28038427 PMCID: PMC5199192 DOI: 10.1016/j.redox.2016.12.021] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 12/19/2016] [Accepted: 12/19/2016] [Indexed: 12/22/2022] Open
Abstract
Activation of hepatic stellate cells (HSCs) is a pivotal event in liver fibrosis, characterized by dramatic disappearance of lipid droplets (LDs). Although LD disappearance has long been considered one of the hallmarks of HSC activation, the underlying molecular mechanisms are largely unknown. In this study, we sought to investigate the role of autophagy in the process of LD disappearance, and to further examine the underlying mechanisms in this molecular context. We found that LD disappearance during HSC activation was associated with a coordinate increase in autophagy. Inhibition or depletion of autophagy by Atg5 siRNA impaired LD disappearance of quiescent HSCs, and also restored lipocyte phenotype of activated HSCs. In contrast, induction of autophagy by Atg5 plasmid accelerated LD loss of quiescent HSCs. Importantly, our study also identified a crucial role for reactive oxygen species (ROS) in the facilitation of autophagy activation. Antioxidants, such as glutathione and N-acetyl cysteine, significantly abrogated ROS production, and in turn, prevented autophagosome generation and autophagic flux during HSC activation. Besides, we found that HSC activation triggered Rab25 overexpression, and promoted the combination of Rab25 and PI3KCIII, which direct autophagy to recognize, wrap and degrade LDs. Down-regulation of Rab25 activity, using Rab25 siRNA, blocked the target recognition of autophagy on LDs, and inhibited LD disappearance of quiescent HSCs. Moreover, the scavenging of excessive ROS could disrupt the interaction between autophagy and Rab25, and increase intracellular lipid content. Overall, these results provide novel implications to reveal the molecular mechanism of LD disappearance during HSC activation, and also identify ROS-Rab25-dependent autophagy as a potential target for the treatment of liver fibrosis. Autophagosome generation and autophagic flux are increased during HSC activation. The inhibition of autophagy blocks LD disappearance of quiescent HSCs. The induction of autophagy accelerates LD disappearance of quiescent HSCs. Rab25 activation is required for autophagy to degrade LDs during HSC activation. Mitochondrial H2O2 production triggers autophagy activation during HSC activation.
Collapse
Affiliation(s)
- Zili Zhang
- Department of Pharmacology, College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Shifeng Zhao
- Department of Pharmacology, College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Zhen Yao
- Department of Pharmacology, College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Ling Wang
- Department of Pharmacology, College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Jiangjuan Shao
- Department of Pharmacy, College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Anping Chen
- Department of Pathology, School of Medicine, Saint Louis University, St Louis., MO 63104, USA
| | - Feng Zhang
- Department of Pharmacology, College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Shizhong Zheng
- Department of Pharmacology, College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, PR China; Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, PR China.
| |
Collapse
|