101
|
Hepatic injury associated with Trypanosoma cruzi infection is attenuated by treatment with 15-deoxy-Δ 12,14 prostaglandin J 2. Exp Parasitol 2016; 170:100-108. [PMID: 27693222 DOI: 10.1016/j.exppara.2016.09.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 09/14/2016] [Accepted: 09/27/2016] [Indexed: 12/19/2022]
Abstract
Trypanosoma cruzi, the etiological agent of Chagas' disease, causes an intense inflammatory response in several tissues, including the liver. Since this organ is central to metabolism, its infection may be reflected in the outcome of the disease. 15-deoxy-Δ12,14 prostaglandin J2 (15dPGJ2), a natural agonist of peroxisome-proliferator activated receptor (PPAR) γ, has been shown to exert anti-inflammatory effects in the heart upon T. cruzi infection. However, its role in the restoration of liver function and reduction of liver inflammation has not been studied yet. BALB/c mice were infected with T. cruzi. The effects of in vivo treatment with 15dPGJ2 on liver inflammation and fibrosis, as well as on the GOT/GPT ratio were studied and the role of NF-κB pathway on 15dPGJ2-mediated effects was analysed. 15dPGJ2 reduced liver inflammatory infiltrates, proinflammatory enzymes and cytokines expression, restored the De Ritis ratio values to normal, reduced the deposits of interstitial and perisinusoidal collagen, reduced the expression of the pro-fibrotic cytokines and inhibited the translocation of the p65 NF-κB subunit to the nucleus. Thus, we showed that 15dPGJ2 is able to significantly reduce the inflammatory response and fibrosis and reduced enzyme markers of liver damage in mice infected with T. cruzi.
Collapse
|
102
|
Panebianco C, Oben JA, Vinciguerra M, Pazienza V. Senescence in hepatic stellate cells as a mechanism of liver fibrosis reversal: a putative synergy between retinoic acid and PPAR-gamma signalings. Clin Exp Med 2016; 17:269-280. [PMID: 27655446 DOI: 10.1007/s10238-016-0438-x] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 09/08/2016] [Indexed: 12/16/2022]
Abstract
Hepatic stellate cells (HSCs), also known as perisinusoidal cells, are pericytes found in the perisinusoidal space of the liver. HSCs are the major cell type involved in liver fibrosis, which is the formation of scar tissue in response to liver damage. When the liver is damaged, stellate cells can shift into an activated state, characterized by proliferation, contractility and chemotaxis. The activated HSCs secrete collagen scar tissue, which can lead to cirrhosis. Recent studies have shown that in vivo activation of HSCs by fibrogenic agents can eventually lead to senescence of these cells, which would contribute to reversal of fibrosis although it may also favor the insurgence of liver cancer. HSCs in their non-active form store huge amounts of retinoic acid derivatives in lipid droplets, which are progressively depleted upon cell activation in injured liver. Retinoic acid is a metabolite of vitamin A (retinol) that mediates the functions of vitamin A, generally required for growth and development. The precise function of retinoic acid and its alterations in HSCs has yet to be elucidated, and nonetheless in various cell types retinoic acid and its receptors (RAR and RXR) are known to act synergistically with peroxisome proliferator-activated receptor gamma (PPAR-gamma) signaling through the activity of transcriptional heterodimers. Here, we review the recent advancements in the understanding of how retinoic acid signaling modulates the fibrogenic potential of HSCs and proposes a synergistic combined action with PPAR-gamma in the reversal of liver fibrosis.
Collapse
Affiliation(s)
- Concetta Panebianco
- Gastroenterology Unit, IRCCS "Casa Sollievo della Sofferenza" Hospital, Viale dei Cappuccini, 1, San Giovanni Rotondo, FG, Italy
| | - Jude A Oben
- Institute for Liver and Digestive Health, Royal Free Hospital, University College London (UCL), London, UK
| | - Manlio Vinciguerra
- Institute for Liver and Digestive Health, Royal Free Hospital, University College London (UCL), London, UK.,Center for Translational Medicine (CTM), International Clinical Research Center (ICRC), St. Anne's University Hospital, Brno, Czech Republic.,Centro Studi Fegato (CSF)-Liver Research Center, Fondazione Italiana Fegato, Trieste, Italy
| | - Valerio Pazienza
- Gastroenterology Unit, IRCCS "Casa Sollievo della Sofferenza" Hospital, Viale dei Cappuccini, 1, San Giovanni Rotondo, FG, Italy.
| |
Collapse
|
103
|
Qiao H, Cao Q, Fu Y, Guan W, Cheng F, Wu J, Jia X, Chen H, Zhou Y. Sex-determining region Y-box 9 acts downstream of NADPH oxidase to influence the effect of leptin on PPARγ1 expression in hepatic stellate cells. Biochim Biophys Acta Mol Basis Dis 2016; 1862:2186-2196. [PMID: 27593694 DOI: 10.1016/j.bbadis.2016.09.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 08/17/2016] [Accepted: 09/01/2016] [Indexed: 12/30/2022]
Abstract
Leptin, an adipocyte-derived hormone, promotes liver fibrogenesis and inhibits the expression of peroxisome-proliferator activated receptor γ (PPARγ), a key transcription factor in inhibition of hepatic stellate cell (HSC) activation, in HSCs. This research aimed to further investigate the mechanisms underlying leptin regulation of PPARγ1 in HSCs in vivo and in vitro. Results demonstrated that sex-determining region Y-box 9 (Sox9) could bind to a site around -2275 within leptin response region of PPARγ1 promoter and inhibited PPARγ1 expression. Sox9 upregulated the expressions of α1(I)collagen and alpha-smooth muscle actin in HSCs. Leptin stimulated Sox9 expression and Sox9 binding to PPARγ1 promoter. The signaling pathways of NADPH oxidase, β-catenin, and delta-like homolog1 (DLK1) mediated leptin upregulation of Sox9 expression. Moreover, there existed crosstalk between NADPH oxidase pathway and β-catenin or DLK1 signaling pathway. Human liver specimens of cirrhosis were shown to be of a large number of the positive HSCs for p47phox (playing a central role in NADPH oxidase activity), 4-hydroxynonenal (a lipid peroxidation product), Sox9, and α-smooth muscle actin whereas PPARγ-positive HSCs were rarely detected. These results might deepen understanding of the molecular mechanisms for leptin inhibition of PPARγ1 expression in HSCs and for the liver fibrosis associated with leptin.
Collapse
Affiliation(s)
- Haowen Qiao
- Department of Biochemistry & Molecular Biology, Medical College, Nantong University, Qi xiou road 19, Nantong 226001, Jiangsu, China
| | - Qing Cao
- Department of Biochemistry & Molecular Biology, Medical College, Nantong University, Qi xiou road 19, Nantong 226001, Jiangsu, China
| | - Yucheng Fu
- The first affiliated hospital of Zhejiang university, Hangzhou 31000, Zhejiang, China
| | - Wei Guan
- Department of Pharmacology, School of Pharmacy, Nantong University, Qi xiou road 19, Nantong 226001, Jiangsu, China
| | - Fangyun Cheng
- Department of Biochemistry & Molecular Biology, Medical College, Nantong University, Qi xiou road 19, Nantong 226001, Jiangsu, China
| | - Juanjuan Wu
- Department of Biochemistry & Molecular Biology, Medical College, Nantong University, Qi xiou road 19, Nantong 226001, Jiangsu, China
| | - Xin Jia
- Department of Biochemistry & Molecular Biology, Medical College, Nantong University, Qi xiou road 19, Nantong 226001, Jiangsu, China
| | - Hongshan Chen
- Department of Biochemistry & Molecular Biology, Medical College, Nantong University, Qi xiou road 19, Nantong 226001, Jiangsu, China
| | - Yajun Zhou
- Department of Biochemistry & Molecular Biology, Medical College, Nantong University, Qi xiou road 19, Nantong 226001, Jiangsu, China.
| |
Collapse
|
104
|
Ezhilarasan D, Evraerts J, Brice S, Buc-Calderon P, Karthikeyan S, Sokal E, Najimi M. Silibinin Inhibits Proliferation and Migration of Human Hepatic Stellate LX-2 Cells. J Clin Exp Hepatol 2016; 6:167-174. [PMID: 27746612 PMCID: PMC5052367 DOI: 10.1016/j.jceh.2016.01.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 01/06/2016] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Proliferation of hepatic stellate cells (HSCs) play pivotal role in the progression of hepatic fibrosis consequent to chronic liver injury. Silibinin (SBN), a flavonoid compound, has shown to possess cell cycle arresting potential against many actively proliferating cancers cell lines. The objective of this study was to evaluate the anti-proliferative and cell cycle arresting properties of SBN in rapidly proliferating human hepatic stellate LX-2 cell line. METHODS LX-2 cells were fed with culture medium supplemented with different concentrations of SBN (10, 50 and 100 μM). After 24 and 96 h of treatment, total cell number was determined by counting. Cytotoxicity was evaluated by trypan blue dye exclusion test. The expression profile of cMyc and peroxisome proliferator-activated receptor-γ (PPAR-γ) protein expressions was evaluated by Western blotting. Oxidative stress marker genes profile was quantified using qPCR. The migratory response of HSCs was observed by scrape wound healing assay. RESULTS SBN treatments significantly inhibit the LX-2 cell proliferation (without affecting its viability) in dose dependent manner. This treatment also retards the migration of LX-2 cells toward injured area. In Western blotting studies SBN treatment up regulated the protein expressions of PPAR-γ and inhibited cMyc. CONCLUSION The present study shows that SBN retards the proliferation, activation and migration of LX-2 cells without inducing cytotoxicity and oxidative stress. The profound effects could be due to cell cycle arresting potential of SBN.
Collapse
Key Words
- AKR1C1, aldo-keto reductase family 1, member C1
- ARE, antioxidant responsive element
- CDKI, cyclin dependent kinase inhibitor
- CYP450, cytochrome P450
- DMEM, Dulbecco's modified Eagle's medium
- DMSO, dimethylsulphoxide
- ECM, extracellular matrix
- FBS, fetal bovine serum
- GAPDH, glyceraldehyde 3-phosphate dehydrogenase
- HMOX1, heme oxygenase (decycling) 1
- HSCs, hepatic stellate cells
- NQO1, NAD(P)H dehydrogenase, quinone 1
- Nrf-2, nuclear respiratory factor
- PPAR-γ, peroxisome proliferator-activated receptor-γ
- PPIA, peptidylprolyl isomerase A
- ROS, reactive oxygen species
- SBN, silibinin
- TXNRD1, thioredoxin reductase 1
- cytotoxicity
- hepatic stellate cells
- oxidative stress
- qPCR, quantitative polymerase chain reaction
- wound healing
Collapse
Affiliation(s)
- Devaraj Ezhilarasan
- Institut de Recherche Expérimentale et Clinique (IREC), Laboratory of Pediatric Hepatology and Cell Therapy, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Jonathan Evraerts
- Institut de Recherche Expérimentale et Clinique (IREC), Laboratory of Pediatric Hepatology and Cell Therapy, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Sid Brice
- Louvain Drug Research Institute, Toxicology and Cancer Biology Research Group, PMNT Unit, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Pedro Buc-Calderon
- Facultad de Ciencias de la Salud, Universidad Arturo Prat, Iquique, Chile
| | - Sivanesan Karthikeyan
- Department of Pharmacology and Environmental Toxicology, Food and Hepatotoxicology Laboratory, Dr. ALM Post Graduate Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Chennai 600 113, India
| | - Etienne Sokal
- Institut de Recherche Expérimentale et Clinique (IREC), Laboratory of Pediatric Hepatology and Cell Therapy, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Mustapha Najimi
- Institut de Recherche Expérimentale et Clinique (IREC), Laboratory of Pediatric Hepatology and Cell Therapy, Université Catholique de Louvain, 1200 Brussels, Belgium,Address for correspondence: Mustapha Najimi, Institut de Recherche Expérimentale & Clinique (IREC), Laboratory of Pediatric Hepatology and Cell Therapy, Avenue Mounier, 52, Box B1.52.03, 1200 Brussels, Belgium.Institut de Recherche Expérimentale & Clinique (IREC), Laboratory of Pediatric Hepatology and Cell TherapyAvenue Mounier, 52, Box B1.52.03Brussels1200Belgium
| |
Collapse
|
105
|
Jang CH, Kim KM, Yang JH, Cho SS, Kim SJ, Shin SM, Cho IJ, Ki SH. The Role of Lipin-1 in the Regulation of Fibrogenesis and TGF-β Signaling in Hepatic Stellate Cells. Toxicol Sci 2016; 153:28-38. [PMID: 27345520 DOI: 10.1093/toxsci/kfw109] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The adipogenic transcriptional regulation was reported to inhibit transdifferentiation of hepatic stellate cells (HSCs), which constitute the main fibrogenic cell type in the liver. Lipin-1 exhibits a dual function: an enzyme that catalyzes the conversion of phosphatidate to diacylglycerol and a transcriptional regulator. However, the involvement of Lipin-1 in the regulation of transforming growth factor-β (TGF-β) signaling and fibrogenesis in HSCs is not fully understood. Here, we showed that Lipin-1 was downregulated in activated primary HSCs and TGF-β-treated LX-2 cells, immortalized human HSC cell lines. The downregulation of Lipin-1 by TGF-β was not dependent on altered mRNA stability but rather on protein stability. Treatment of LX-2 cells with the proteasome inhibitor led to the accumulation of Lipin-1. Moreover, we observed a significant increase in Lipin-1 polyubiquitination. Overexpression of Lipin-1 attenuated TGF-β-induced fibrogenic gene expression. In addition, Lipin-1 inhibited TGF-β-mediated activation of Sma and Mad-related family (SMAD), a major transcription factor that transduces intracellular signals from TGF-β. Resveratrol, a well-known natural polyphenolic antioxidant, is known to inhibit liver fibrosis, although its mechanism of action remains unknown. Our data showed that resveratrol significantly increased the levels of Lipin-1 protein and mRNA in HSCs. Further investigation revealed that resveratrol blocked the polyubiquitination of Lipin-1. Resveratrol inhibited TGF-β-induced fibrogenic gene expression. TGF-β-induced SMAD binding element-luciferase reporter activity was significantly diminished by resveratrol with a simultaneous decrease in SMAD3 phosphorylation. Consistently, knockdown of the Lipin-1 gene using siRNA abolished the inhibitory effect of resveratrol. We conclude that Lipin-1 can antagonize HSC activation through the inhibition of TGF-β/SMAD signaling and that resveratrol may affect Lipin-1 gene induction and contribute to the inhibition of TGF-β-mediated hepatic fibrogenesis.
Collapse
Affiliation(s)
- Chang Ho Jang
- *College of Pharmacy, Chosun University, Gwangju, 61452, Korea
| | - Kyu Min Kim
- *College of Pharmacy, Chosun University, Gwangju, 61452, Korea College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Korea
| | - Ji Hye Yang
- *College of Pharmacy, Chosun University, Gwangju, 61452, Korea
| | - Sam Seok Cho
- *College of Pharmacy, Chosun University, Gwangju, 61452, Korea
| | - Seung Jung Kim
- *College of Pharmacy, Chosun University, Gwangju, 61452, Korea
| | - Sang Mi Shin
- *College of Pharmacy, Chosun University, Gwangju, 61452, Korea
| | - Il Je Cho
- MRC-GHF, College of Korean Medicine, Daegu Haany University, Gyeongsan, 38610, Korea
| | - Sung Hwan Ki
- *College of Pharmacy, Chosun University, Gwangju, 61452, Korea
| |
Collapse
|
106
|
de Souza ICC, Martins LAM, de Vasconcelos M, de Oliveira CM, Barbé-Tuana F, Andrade CB, Pettenuzzo LF, Borojevic R, Margis R, Guaragna R, Guma FCR. Resveratrol Regulates the Quiescence-Like Induction of Activated Stellate Cells by Modulating the PPARγ/SIRT1 Ratio. J Cell Biochem 2016; 116:2304-12. [PMID: 25833683 DOI: 10.1002/jcb.25181] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 03/31/2015] [Indexed: 01/27/2023]
Abstract
The activation of hepatic stellate cell (HSC), from a quiescent cell featuring cytoplasmic lipid droplets to a proliferative myofibroblast, plays an important role in liver fibrosis development. The GRX line is an activated HSC model that can be induced by all-trans-retinol to accumulate lipid droplets. Resveratrol is known for activating Sirtuin1 (SIRT1), a NAD(+)-dependent deacetylase that suppresses the activity of peroxisome proliferator-activated receptor gamma (PPARγ), an important adipogenic transcription factor involved in the quiescence maintenance of HSC. We evaluated the effects of 0.1 μM of resveratrol in retinol-induced GRX quiescence by investigating the interference of SIRT1 and PPARγ on cell lipogenesis. GRX lipid accumulation was evaluated through Oil-red O staining, triacylglycerides quantification, and [(14)C] acetate incorporation into lipids. mRNA expression and protein content of SIRT1 and PPARγ were measured by RT-PCR and immunoblotting, respectively. Resveratrol-mediated SIRT1 stimuli did not induce lipogenesis and reduced the retinol-mediated fat-storing capacity in GRX. In order to support our results, we established a cell culture model of transgenic super expression of PPARγ in GRX cells (GRXPγ). Resveratrol reduced lipid droplets accumulation in GRXPγ cells. These results suggest that the PPARγ/SIRT1 ratio plays an important role in the fate of HSC. Thus, whenever the PPARγ activity is greater than SIRT1 activity the lipogenesis is enabled.
Collapse
Affiliation(s)
- Izabel Cristina Custódio de Souza
- Departamento de Morfologia, IB, Universidade Federal de Pelotas (UFPel), av. Duque de Caxias, 250, CEP 96 030 000, Pelotas, RS, Brazil.,Departamento de Bioquímica, ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), rua Ramiro Barcelos, 2600-Anexo I, CEP 90035-003, Porto Alegre, RS, Brazil
| | - Leo Anderson Meira Martins
- Departamento de Bioquímica, ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), rua Ramiro Barcelos, 2600-Anexo I, CEP 90035-003, Porto Alegre, RS, Brazil
| | - Mariana de Vasconcelos
- Departamento de Bioquímica, ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), rua Ramiro Barcelos, 2600-Anexo I, CEP 90035-003, Porto Alegre, RS, Brazil
| | - Cleverson Moraes de Oliveira
- Departamento de Bioquímica, ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), rua Ramiro Barcelos, 2600-Anexo I, CEP 90035-003, Porto Alegre, RS, Brazil
| | - Florencia Barbé-Tuana
- Departamento de Bioquímica, ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), rua Ramiro Barcelos, 2600-Anexo I, CEP 90035-003, Porto Alegre, RS, Brazil
| | | | - Letícia Ferreira Pettenuzzo
- Departamento de Bioquímica, ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), rua Ramiro Barcelos, 2600-Anexo I, CEP 90035-003, Porto Alegre, RS, Brazil
| | - Radovan Borojevic
- Departamento de Histologia e Embriologia, ICB, PABCAM, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | - Rogério Margis
- Departamento de Biofísica, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Regina Guaragna
- Departamento de Bioquímica, ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), rua Ramiro Barcelos, 2600-Anexo I, CEP 90035-003, Porto Alegre, RS, Brazil
| | - Fátima Costa Rodrigues Guma
- Departamento de Bioquímica, ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), rua Ramiro Barcelos, 2600-Anexo I, CEP 90035-003, Porto Alegre, RS, Brazil
| |
Collapse
|
107
|
van der Veen JN, Lingrell S, Gao X, Quiroga AD, Takawale A, Armstrong EA, Yager JY, Kassiri Z, Lehner R, Vance DE, Jacobs RL. Pioglitazone attenuates hepatic inflammation and fibrosis in phosphatidylethanolamine N-methyltransferase-deficient mice. Am J Physiol Gastrointest Liver Physiol 2016; 310:G526-38. [PMID: 26797396 DOI: 10.1152/ajpgi.00243.2015] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 12/21/2015] [Indexed: 01/31/2023]
Abstract
Phosphatidylethanolamine N-methyltransferase (PEMT) is an important enzyme in hepatic phosphatidylcholine (PC) biosynthesis. Pemt(-/-) mice are protected against high-fat diet (HFD)-induced obesity and insulin resistance; however, these mice develop nonalcoholic fatty liver disease (NAFLD). We hypothesized that peroxisomal proliferator-activated receptor-γ (PPARγ) activation by pioglitazone might stimulate adipocyte proliferation, thereby directing lipids from the liver toward white adipose tissue. Pioglitazone might also act directly on PPARγ in the liver to improve NAFLD. Pemt(+/+) and Pemt(-/-) mice were fed a HFD with or without pioglitazone (20 mg·kg(-1)·day(-1)) for 10 wk. Pemt(-/-) mice were protected from HFD-induced obesity but developed NAFLD. Treatment with pioglitazone caused an increase in body weight gain in Pemt(-/-) mice that was mainly due to increased adiposity. Moreover, pioglitazone improved NAFLD in Pemt(-/-) mice, as indicated by a 35% reduction in liver weight and a 57% decrease in plasma alanine transaminase levels. Livers from HFD-fed Pemt(-/-) mice were steatotic, inflamed, and fibrotic. Hepatic steatosis was still evident in pioglitazone-treated Pemt(-/-) mice; however, treatment with pioglitazone reduced hepatic fibrosis, as evidenced by reduced Sirius red staining and lowered mRNA levels of collagen type Iα1 (Col1a1), tissue inhibitor of metalloproteinases 1 (Timp1), α-smooth muscle actin (Acta2), and transforming growth factor-β (Tgf-β). Similarly, oxidative stress and inflammation were reduced in livers from Pemt(-/-) mice upon treatment with pioglitazone. Together, these data show that activation of PPARγ in HFD-fed Pemt(-/-) mice improved liver function, while these mice were still protected against diet-induced obesity and insulin resistance.
Collapse
Affiliation(s)
- Jelske N van der Veen
- Group on the Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, Alberta, Canada; Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Susanne Lingrell
- Group on the Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, Alberta, Canada; Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Xia Gao
- Group on the Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, Alberta, Canada; Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Ariel D Quiroga
- Group on the Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, Alberta, Canada; Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Abhijit Takawale
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada; and
| | - Edward A Armstrong
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Jerome Y Yager
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Zamaneh Kassiri
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada; and
| | - Richard Lehner
- Group on the Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, Alberta, Canada; Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Dennis E Vance
- Group on the Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, Alberta, Canada; Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | - René L Jacobs
- Group on the Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, Alberta, Canada; Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
108
|
Nati M, Haddad D, Birkenfeld AL, Koch CA, Chavakis T, Chatzigeorgiou A. The role of immune cells in metabolism-related liver inflammation and development of non-alcoholic steatohepatitis (NASH). Rev Endocr Metab Disord 2016; 17:29-39. [PMID: 26847547 DOI: 10.1007/s11154-016-9339-2] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The low grade inflammatory state present in obesity promotes the progression of Non-Alcoholic Fatty Liver Disease (NAFLD). In Non-Alcoholic Steatohepatitis (NASH), augmented hepatic steatosis is accompanied by aberrant intrahepatic inflammation and exacerbated hepatocellular injury. NASH is an important disorder and can lead to fibrosis, cirrhosis and even neoplasia. The pathology of NASH involves a complex network of mechanisms, including increased infiltration of different subsets of immune cells, such as monocytes, T-lymphocytes and neutrophils, to the liver, as well as activation and in situ expansion of liver resident cells such as Kupffer cells or stellate cells. In this review, we summarize recent advances regarding understanding the role of the various cells of the innate and adaptive immunity in NASH development and progression, and discuss possible future therapeutic options and tools to interfere with disease progression.
Collapse
Affiliation(s)
- Marina Nati
- Department of Clinical Pathobiochemistry, Faculty of Medicine, Technische Universität Dresden, MTZ, Fiedlerstrasse 42, 01307, Dresden, Germany
| | - David Haddad
- Department of Clinical Pathobiochemistry, Faculty of Medicine, Technische Universität Dresden, MTZ, Fiedlerstrasse 42, 01307, Dresden, Germany
| | - Andreas L Birkenfeld
- Section of Metabolic Vascular Medicine, Medical Clinic III, Faculty of Medicine, TU Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine, TU Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
- Division of Diabetes and Nutritional Sciences, Rayne Institute, King's College London, London, UK
| | - Christian A Koch
- Division of Endocrinology, Endocrine Tumor Program, Cancer Institute, University of Mississippi Medical Center, Jackson, MS, USA
| | - Triantafyllos Chavakis
- Department of Clinical Pathobiochemistry, Faculty of Medicine, Technische Universität Dresden, MTZ, Fiedlerstrasse 42, 01307, Dresden, Germany
- Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine, TU Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, Dresden, Germany
| | - Antonios Chatzigeorgiou
- Department of Clinical Pathobiochemistry, Faculty of Medicine, Technische Universität Dresden, MTZ, Fiedlerstrasse 42, 01307, Dresden, Germany.
- Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine, TU Dresden, Dresden, Germany.
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany.
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, TU Dresden, Dresden, Germany.
| |
Collapse
|
109
|
Oró D, Yudina T, Fernández-Varo G, Casals E, Reichenbach V, Casals G, González de la Presa B, Sandalinas S, Carvajal S, Puntes V, Jiménez W. Cerium oxide nanoparticles reduce steatosis, portal hypertension and display anti-inflammatory properties in rats with liver fibrosis. J Hepatol 2016; 64:691-8. [PMID: 26519601 DOI: 10.1016/j.jhep.2015.10.020] [Citation(s) in RCA: 141] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 10/01/2015] [Accepted: 10/23/2015] [Indexed: 12/31/2022]
Abstract
BACKGROUND & AIMS Cerium oxide nanoparticles (CeO2NPs) have proven to behave as free radical scavengers and/or anti-inflammatory agents. The aim of the study was to determine whether CeO2NPs display hepatoprotective properties in experimental chronic liver disease. METHODS Systemic and hepatic effects of nanoparticles were assessed in CCl4-treated rats receiving CeO2NPs or vehicle twice weekly for two weeks and CCl4 treatment was continued for 8 additional weeks. Thereafter, mean arterial pressure and portal pressure (PP) were assessed and serum samples obtained to measure standard hepatic and renal function tests. Organ and subcellular distribution of NPs were assessed using mass spectrometry (ICP-MS) and transmission electron microscopy. Liver samples were obtained to evaluate steatosis, α-SMA expression, macrophage infiltration, apoptosis and mRNA expression of oxidative stress, inflammatory or vasoactive related genes. RESULTS Most CeO2NPs were located in the liver and it reduced hepatic steatosis, ameliorated systemic inflammatory biomarkers and improved PP without affecting mean arterial pressure. In addition, a marked reduction in mRNA expression of inflammatory cytokines (TNFα, IL1β, COX-2, iNOS), ET-1 and messengers related to oxidative (Epx, Ncf1, Ncf2) or endoplasmic reticulum (Atf3, Hspa5) stress signaling pathways was observed in the liver of rats receiving CeO2NPs. This was associated with reduced macrophage infiltration and reduced abundance of caspase-3, α-SMA and inflammatory cytokines. CONCLUSIONS CeO2NPs administration to CCl4-treated rats protects against chronic liver injury by reducing liver steatosis and portal hypertension and markedly attenuating the intensity of the inflammatory response, thereby suggesting that CeO2NPs may be of therapeutic value in chronic liver disease.
Collapse
Affiliation(s)
- Denise Oró
- Biochemistry and Molecular Genetics Service, Hospital Clínic Universitari, IDIBAPS, CIBERehd, Barcelona, Spain
| | - Tetyana Yudina
- Institut Català de Nanociència i Nanotecnologia (ICN2), Bellaterra, Spain
| | - Guillermo Fernández-Varo
- Biochemistry and Molecular Genetics Service, Hospital Clínic Universitari, IDIBAPS, CIBERehd, Barcelona, Spain; Department Ciencies Fisiologiques I, University of Barcelona, Barcelona, Spain
| | - Eudald Casals
- Institut Català de Nanociència i Nanotecnologia (ICN2), Bellaterra, Spain
| | - Vedrana Reichenbach
- Biochemistry and Molecular Genetics Service, Hospital Clínic Universitari, IDIBAPS, CIBERehd, Barcelona, Spain
| | - Gregori Casals
- Biochemistry and Molecular Genetics Service, Hospital Clínic Universitari, IDIBAPS, CIBERehd, Barcelona, Spain
| | | | - Silvia Sandalinas
- Biochemistry and Molecular Genetics Service, Hospital Clínic Universitari, IDIBAPS, CIBERehd, Barcelona, Spain
| | - Silvia Carvajal
- Biochemistry and Molecular Genetics Service, Hospital Clínic Universitari, IDIBAPS, CIBERehd, Barcelona, Spain
| | - Victor Puntes
- Institut Català de Nanociència i Nanotecnologia (ICN2), Bellaterra, Spain; Institut Català de Recerca i Estudis Avançats, (ICREA), Barcelona, Spain; Vall d'Hebron Insitute of Research (VHIR), Barcelona, Spain.
| | - Wladimiro Jiménez
- Biochemistry and Molecular Genetics Service, Hospital Clínic Universitari, IDIBAPS, CIBERehd, Barcelona, Spain; Department Ciencies Fisiologiques I, University of Barcelona, Barcelona, Spain.
| |
Collapse
|
110
|
Abstract
BACKGROUND Intestinal fibrosis is mainly associated with Crohn's disease and is defined as a progressive and excessive deposition of extracellular matrix components. No specific antifibrotic therapies are available. In this study, we evaluate the antifibrotic effect of a novel 5-ASA analog able to activate the peroxisome proliferator-activated receptor γ, named GED-0507-34 Levo. METHODS Colonic fibrosis was induced in 110 C57BL/6 mice by 3 cycles of 2.5% (wt/vol) dextran sulfate sodium administration for 6 weeks. The preventive effects of oral daily GED (30 mg · kg(-1) · d(-1)) administration were evaluated using a macroscopic and histological score and also through biological endpoints. Expression of main markers of myofibroblasts activation was determined in transforming growth factor (TGF-β)-stimulated intestinal fibroblasts and epithelial cells. RESULTS GED improved macroscopic and microscopic intestinal lesions in dextran sulfate sodium-treated animals and reduced the profibrotic gene expression of Acta2, COL1a1, and Fn1 by 1.48-folds (P < 0.05), 1.93-folds (P < 0.005), and 1.03-fold (P < 0.05), respectively. It reduced protein levels of main markers of fibrosis (α-SMA and Collagen I-II) and the main TGF-β/Smad pathway components. GED also decreased the interleukin-13 and connective tissue growth factor expression by 1.89-folds (P < 0.05) and 2.2-folds (P < 0.005), respectively. GED inhibited TGF-β-induced activation of both fibroblast and intestinal epithelial cell lines, by regulating mRNA expression of α-SMA and fibronectin, and restoring the TGF-β-induced loss of intestinal epithelial cell markers. GED treatment also reduced the TGF-β and ACTA1 expression in primary human intestinal fibroblasts from ulcerative colitis patients. CONCLUSIONS GED ameliorates intestinal fibrosis in dextran sulfate sodium-induced chronic colitis in mice and regulates major profibrotic cellular and molecular mechanisms.
Collapse
|
111
|
Floreani A, Franceschet I, Perini L, Cazzagon N, Gershwin ME, Bowlus CL. New therapies for primary biliary cirrhosis. Clin Rev Allergy Immunol 2016; 48:263-72. [PMID: 25331740 DOI: 10.1007/s12016-014-8456-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Primary biliary cirrhosis (PBC) is a rare inflammatory liver disease for which ursodeoxycholic acid (UDCA) is the only therapy approved by the U.S. Food and Drug Administration. Patients with a biochemical response to UDCA therapy have a similar survival rate compared to the general population. However, up to 40% of PBC patients do not achieve a complete response to UDCA, have an increased risk of liver-related death and liver transplantation, and represent a persistent medical need for new therapies. Several novel drugs have recently been studied and show potential efficacy in PBC. Obeticholic acid, a farnesoid X receptor agonist, has been tested in phase II trials and initial results after 1 year in a phase III international trial suggest that it may be effective in achieving a biochemical response in approximately 40% of patients who do not completely respond to UDCA. Several small studies on fibrates have suggested that they may have efficacy, but larger studies are needed. Surprisingly, results of immunomodulators and biologics have not yet been able to demonstrate efficacy, but new approaches have shown promise in animal models and their translation to human clinical trials are awaited.
Collapse
Affiliation(s)
- Annarosa Floreani
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy,
| | | | | | | | | | | |
Collapse
|
112
|
Zhou X, Zang X, Ponnusamy M, Masucci MV, Tolbert E, Gong R, Zhao TC, Liu N, Bayliss G, Dworkin LD, Zhuang S. Enhancer of Zeste Homolog 2 Inhibition Attenuates Renal Fibrosis by Maintaining Smad7 and Phosphatase and Tensin Homolog Expression. J Am Soc Nephrol 2015; 27:2092-108. [PMID: 26701983 DOI: 10.1681/asn.2015040457] [Citation(s) in RCA: 133] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 10/30/2015] [Indexed: 01/06/2023] Open
Abstract
Enhancer of zeste homolog 2 (EZH2) is a methyltransferase that induces histone H3 lysine 27 trimethylation (H3K27me3) and functions as an oncogenic factor in many cancer types. However, the role of EZH2 in renal fibrogenesis remains unexplored. In this study, we found high expression of EZH2 and H3K27me3 in cultured renal fibroblasts and fibrotic kidneys from mice with unilateral ureteral obstruction and humans with CKD. Pharmacologic inhibition of EZH2 with 3-deazaneplanocin A (3-DZNeP) or GSK126 or siRNA-mediated silencing of EZH2 inhibited serum- and TGFβ1-induced activation of renal interstitial fibroblasts in vitro, and 3-DZNeP administration abrogated deposition of extracellular matrix proteins and expression of α-smooth muscle actin in the obstructed kidney. Injury to the kidney enhanced Smad7 degradation, Smad3 phosphorylation, and TGFβ receptor 1 expression, and 3-DZNeP administration prevented these effects. 3-DZNeP also suppressed phosphorylation of the renal EGF and PDGFβ receptors and downstream signaling molecules signal transducer and activator of transcription 3 and extracellular signal-regulated kinase 1/2 after injury. Moreover, EZH2 inhibition increased the expression of phosphatase and tensin homolog (PTEN), a protein previously associated with dephosphorylation of tyrosine kinase receptors in the injured kidney and serum-stimulated renal interstitial fibroblasts. Finally, blocking PTEN with SF1670 largely diminished the inhibitory effect of 3-DZNeP on renal myofibroblast activation. These results uncovered the important role of EZH2 in mediating the development of renal fibrosis by downregulating expression of Smad7 and PTEN, thus activating profibrotic signaling pathways. Targeted inhibition of EZH2, therefore, could be a novel therapy for treating CKD.
Collapse
Affiliation(s)
- Xiaoxu Zhou
- Department of Medicine, Rhode Island Hospital and Alpert Medical School, Brown University, Providence, Rhode Island; Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiujuan Zang
- Department of Medicine, Rhode Island Hospital and Alpert Medical School, Brown University, Providence, Rhode Island; Department of Nephrology, Shanghai Songjiang District Central Hospital, Shanghai, China
| | - Murugavel Ponnusamy
- Department of Medicine, Rhode Island Hospital and Alpert Medical School, Brown University, Providence, Rhode Island
| | - Monica V Masucci
- Department of Medicine, Rhode Island Hospital and Alpert Medical School, Brown University, Providence, Rhode Island
| | - Evelyn Tolbert
- Department of Medicine, Rhode Island Hospital and Alpert Medical School, Brown University, Providence, Rhode Island
| | - Rujun Gong
- Department of Medicine, Rhode Island Hospital and Alpert Medical School, Brown University, Providence, Rhode Island
| | - Ting C Zhao
- Department of Surgery, Boston University Medical School, Roger Williams Medical Center, Boston University, Providence, Rhode Island; and
| | - Na Liu
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - George Bayliss
- Department of Medicine, Rhode Island Hospital and Alpert Medical School, Brown University, Providence, Rhode Island
| | - Lance D Dworkin
- Department of Medicine, Rhode Island Hospital and Alpert Medical School, Brown University, Providence, Rhode Island
| | - Shougang Zhuang
- Department of Medicine, Rhode Island Hospital and Alpert Medical School, Brown University, Providence, Rhode Island; Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
113
|
Zheng Z, Zhang X, Wang J, Dandekar A, Kim H, Qiu Y, Xu X, Cui Y, Wang A, Chen LC, Rajagopalan S, Sun Q, Zhang K. Exposure to fine airborne particulate matters induces hepatic fibrosis in murine models. J Hepatol 2015; 63:1397-404. [PMID: 26220751 PMCID: PMC5003300 DOI: 10.1016/j.jhep.2015.07.020] [Citation(s) in RCA: 129] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Revised: 07/07/2015] [Accepted: 07/16/2015] [Indexed: 01/13/2023]
Abstract
BACKGROUND & AIMS Hepatic fibrosis, featured by the accumulation of excessive extracellular matrix in liver tissue, is associated with metabolic disease and cancer. Inhalation exposure to airborne particulate matter in fine ranges (PM2.5) correlates with pulmonary dysfunction, cardiovascular disease, and metabolic syndrome. In this study, we investigated the effect and mechanism of PM2.5 exposure on hepatic fibrogenesis. METHODS Both inhalation exposure of mice and in vitro exposure of specialized cells to PM2.5 were performed to elucidate the effect of PM2.5 exposure on hepatic fibrosis. Histological examinations, gene expression analyses, and genetic animal models were utilized to determine the effect and mechanism by which PM2.5 exposure promotes hepatic fibrosis. RESULTS Inhalation exposure to concentrated ambient PM2.5 induces hepatic fibrosis in mice under the normal chow or high-fat diet. Mice after PM2.5 exposure displayed increased expression of collagens in liver tissues. Exposure to PM2.5 led to activation of the transforming growth factor β-SMAD3 signaling, suppression of peroxisome proliferator-activated receptor γ, and expression of collagens in hepatic stellate cells. NADPH oxidase plays a critical role in PM2.5-induced liver fibrogenesis. CONCLUSIONS Exposure to PM2.5 exerts discernible effects on promoting hepatic fibrogenesis. NADPH oxidase mediates the effects of PM2.5 exposure on promoting hepatic fibrosis.
Collapse
Affiliation(s)
- Ze Zheng
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Xuebao Zhang
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Jiemei Wang
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Aditya Dandekar
- Department of Immunology and Microbiology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Hyunbae Kim
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Yining Qiu
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Xiaohua Xu
- Division of Environmental Health Sciences, College of Public Health, Ohio State University, Columbus, OH 43210, USA
| | - Yuqi Cui
- Division of Cardiovascular Medicine, Davis Heart & Lung Research Institute, College of Medicine, Ohio State University, Columbus, OH 43210, USA
| | - Aixia Wang
- Division of Cardiovascular Medicine, Davis Heart & Lung Research Institute, College of Medicine, Ohio State University, Columbus, OH 43210, USA; Division of Environmental Health Sciences, College of Public Health, Ohio State University, Columbus, OH 43210, USA
| | - Lung Chi Chen
- Department of Environmental Medicine, New York University, Tuxedo, NY 10987, USA
| | - Sanjay Rajagopalan
- Division of Cardiovascular Medicine, Davis Heart & Lung Research Institute, College of Medicine, Ohio State University, Columbus, OH 43210, USA
| | - Qinghua Sun
- Division of Cardiovascular Medicine, Davis Heart & Lung Research Institute, College of Medicine, Ohio State University, Columbus, OH 43210, USA; Division of Environmental Health Sciences, College of Public Health, Ohio State University, Columbus, OH 43210, USA
| | - Kezhong Zhang
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA; Department of Immunology and Microbiology, Wayne State University School of Medicine, Detroit, MI 48201, USA.
| |
Collapse
|
114
|
Wang W, Yan M, Ji Q, Lu J, Ji Y, Ji J. Suberoylanilide hydroxamic acid suppresses hepatic stellate cells activation by HMGB1 dependent reduction of NF-κB1. PeerJ 2015; 3:e1362. [PMID: 26557438 PMCID: PMC4636417 DOI: 10.7717/peerj.1362] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 10/08/2015] [Indexed: 01/20/2023] Open
Abstract
Hepatic stellate cells (HSCs) activation is essential to the pathogenesis of liver fibrosis. Exploring drugs targeting HSC activation is a promising anti-fibrotic strategy. In the present study, we found suberoylanilide hydroxamic acid (SAHA), a histone deacetylase inhibitor, prominently suppressed the activation phenotype of a human hepatic stellate cell line—LX2. The production of collagen type I and α-smooth muscle actin (α-SMA) as well as the proliferation and migration of LX2 cells were significantly reduced by SAHA treatment. To determine the molecular mechanisms underlying this suppression, genome wild gene regulation by SAHA was determined by Affymetrix 1.0 human cDNA array. Upon SAHA treatment, the abundance of 331 genes was up-regulated and 173 genes was down-regulated in LX2 cells. Bioinformatic analyses of these altered genes highlighted the high mobility group box 1 (HMGB1) pathway was one of the most relevant pathways that contributed to SAHA induced suppression of HSCs activation. Further studies demonstrated the increased acetylation of intracellular HMGB1 in SAHA treated HSCs, and this increasing is most likely to be responsible for SAHA induced down-regulation of nuclear factor kappa B1 (NF-κB1) and is one of the main underlying mechanisms for the therapeutic effect of SAHA for liver fibrosis.
Collapse
Affiliation(s)
- Wenwen Wang
- Department of Pathology, Medical School of Nantong University , Nantong , China ; Department of Pathology, Traditional Chinese Medicine Hospital of Jiangyin City , Jiangyin , China
| | - Min Yan
- Department of Pathology, Medical School of Nantong University , Nantong , China
| | - Qiuhong Ji
- Neurology Department, Affiliated Hospital of Nantong University , Nantong , China
| | - Jinbiao Lu
- Department of Pathology, Medical School of Nantong University , Nantong , China
| | - Yuhua Ji
- Key Laboratory of Neuroregeneration, Nantong University , Nantong , China
| | - Juling Ji
- Department of Pathology, Medical School of Nantong University , Nantong , China
| |
Collapse
|
115
|
Zhao ZH, Fan YC, Zhao Q, Dou CY, Ji XF, Zhao J, Gao S, Li XY, Wang K. Promoter methylation status and expression of PPAR-γ gene are associated with prognosis of acute-on-chronic hepatitis B liver failure. Clin Epigenetics 2015; 7:115. [PMID: 26516376 PMCID: PMC4625884 DOI: 10.1186/s13148-015-0149-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 10/14/2015] [Indexed: 12/12/2022] Open
Abstract
Background Peroxisome proliferator-activated receptor gamma (PPAR-γ) has been demonstrated to be involved in anti-inflammatory reactions, but its role in acute-on-chronic hepatitis B liver failure (ACHBLF) is unclear. Therefore, DNA methylation patterns and expression level of PPAR-γ gene were detected in peripheral blood mononuclear cells (PBMCs) from 81 patients with ACHBLF, 50 patients with chronic hepatitis B (CHB), and 30 healthy controls, and the possible role of PPAR-γ in ACHBLF was analyzed. Results We found that aberrant PPAR-γ promoter methylation was attenuated in ACHBLF patients compared with CHB patients and was responsible for the elevated PPAR-γ expression level, which was negatively correlated with total bilirubin and international normalized ratio. Plasma level of TNF-α and IL-6 in ACHBLF patients were higher than CHB patients and healthy controls and significantly reduced in unmethylated group. ACHBLF patients with PPAR-γ promoter methylation had poorer outcomes than those without. Correspondingly, PPAR-γ messenger RNA (mRNA) level was higher in survivors than non-survivors and gradually increased in survivors with time, while remained low level in non-survivors. Conclusions Aberrant promoter methylation decline and PPAR-γ expression rebound occurred in ACHBLF compared with CHB and could improve prognosis of ACHBLF by negatively regulating cytokines. Electronic supplementary material The online version of this article (doi:10.1186/s13148-015-0149-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ze-Hua Zhao
- Department of Hepatology, Qilu Hospital of Shandong University, Wenhuaxi Road 107#, Jinan, 250012 China
| | - Yu-Chen Fan
- Department of Hepatology, Qilu Hospital of Shandong University, Wenhuaxi Road 107#, Jinan, 250012 China ; Institute of Hepatology, Shandong University, Wenhuaxi Road 107#, Jinan, 250012 China
| | - Qi Zhao
- Department of Gastroenterology, Provincial Hospital Affiliated to Shandong University, Jinan, 250012 China
| | - Cheng-Yun Dou
- Department of Hepatology, Qilu Hospital of Shandong University, Wenhuaxi Road 107#, Jinan, 250012 China
| | - Xiang-Fen Ji
- Department of Hepatology, Qilu Hospital of Shandong University, Wenhuaxi Road 107#, Jinan, 250012 China
| | - Jing Zhao
- Department of Hepatology, Qilu Hospital of Shandong University, Wenhuaxi Road 107#, Jinan, 250012 China
| | - Shuai Gao
- Department of Hepatology, Qilu Hospital of Shandong University, Wenhuaxi Road 107#, Jinan, 250012 China
| | - Xin-You Li
- Department of Hepatology, Qilu Hospital of Shandong University, Wenhuaxi Road 107#, Jinan, 250012 China
| | - Kai Wang
- Department of Hepatology, Qilu Hospital of Shandong University, Wenhuaxi Road 107#, Jinan, 250012 China ; Institute of Hepatology, Shandong University, Wenhuaxi Road 107#, Jinan, 250012 China
| |
Collapse
|
116
|
Abstract
Recent research on hepatic stellate cells (HSCs) has spotlighted the involvement of morphogens in their cell fate determination in liver fibrosis. Temporally and spatially expressed during embryonic development, morphogens are involved in regulation of cell proliferation and differentiation, and tissue patterning. In normal adult liver, morphogens are generally expressed at low levels. However, in liver disease, myofibroblastic HSCs express morphogens such as Wnt, Shh, Necdin, DLK1, and Notch as part of their participation in fibrogenesis and wound healing. Liver regeneration involves cell proliferation and differentiation akin to embryonic liver development where the cells appear to undergo similar fates, and not surprisingly the morphogens are re-activated for the regenerative purpose in adult liver injury. Evidence also points to crosstalk of these morphogens in regulation of HSC fate determination. Genetic ablation or pharmacologic inhibition of morphogens reverts activated HSC to quiescent cells in culture and attenuates progression of hepatic fibrosis. However, positive regulation of liver regeneration by the morphogens needs to be spared. Therapeutically, manipulation of morphogen activities in a cell type and phase-specific manner should offer new modalities for chronic liver disease.
Collapse
|
117
|
El Taghdouini A, Najimi M, Sancho-Bru P, Sokal E, van Grunsven LA. In vitro reversion of activated primary human hepatic stellate cells. FIBROGENESIS & TISSUE REPAIR 2015; 8:14. [PMID: 26251672 PMCID: PMC4527231 DOI: 10.1186/s13069-015-0031-z] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 07/15/2015] [Indexed: 01/10/2023]
Abstract
Background Liver fibrosis is characterized by the excessive formation and accumulation of matrix proteins as a result of wound healing in the liver. A main event during fibrogenesis is the activation of the liver resident quiescent hepatic stellate cell (qHSC). Recent studies suggest that reversion of the activated HSC (aHSC) phenotype into a quiescent-like phenotype could be a major cellular mechanism underlying fibrosis regression in the liver, thereby offering new therapeutic perspectives for the treatment of liver fibrosis. Whether human HSCs have the ability to undergo a similar reversion in phenotype is currently unknown. The aim of the present study is to identify experimental conditions that can revert the in vitro activated phenotype of primary human HSCs and consequently to map the molecular events associated with this reversion process by gene expression profiling. Results We find that epidermal growth factor (EGF) and fibroblast growth factor 2 (FGF2) synergistically downregulate the expression of ACTA2 and LOX in primary human aHSCs. Their combination with oleic acid, palmitic acid, and retinol further potentiates a more quiescent-like phenotype as demonstrated by the abundant presence of retinyl ester-positive intra-cytoplasmic lipid droplets, low expression levels of activation markers, and a reduced basal as well as cytokine-stimulated proliferation and matrix metalloproteinase activity. Gene expression profiling experiments reveal that these in vitro reverted primary human HSCs (rHSCs) display an intermediary phenotype that is distinct from qHSCs and aHSCs. Interestingly, this intermediary phenotype is characterized by the increased expression of several previously identified signature genes of in vivo inactivated mouse HSCs such as CXCL1, CXCL2, and CTSS, suggesting also a potential role for these genes in promoting a quiescent-like phenotype in human HSCs. Conclusions We provide evidence for the ability of human primary aHSCs to revert in vitro to a transitional state through synergistic action of EGF, FGF2, dietary fatty acids and retinol, and provide a first phenotypic and genomic characterization of human in vitro rHSCs. Electronic supplementary material The online version of this article (doi:10.1186/s13069-015-0031-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Adil El Taghdouini
- Liver Cell Biology Lab, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Mustapha Najimi
- Laboratory of Pediatric Hepatology and Cell Therapy, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium
| | - Pau Sancho-Bru
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Etienne Sokal
- Laboratory of Pediatric Hepatology and Cell Therapy, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium
| | - Leo A van Grunsven
- Liver Cell Biology Lab, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussels, Belgium
| |
Collapse
|
118
|
Povero D, Panera N, Eguchi A, Johnson CD, Papouchado BG, de Araujo Horcel L, Pinatel EM, Alisi A, Nobili V, Feldstein AE. Lipid-induced hepatocyte-derived extracellular vesicles regulate hepatic stellate cell via microRNAs targeting PPAR-γ. Cell Mol Gastroenterol Hepatol 2015; 1:646-663.e4. [PMID: 26783552 PMCID: PMC4714359 DOI: 10.1016/j.jcmgh.2015.07.007] [Citation(s) in RCA: 159] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND&AIMS Hepatic stellate cells (HSCs) play a key role in liver fibrosis in various chronic liver disorders including nonalcoholic fatty liver disease (NAFLD). The development of liver fibrosis requires a phenotypic switch from quiescent to activated HSCs. The triggers for HSCs activation in NAFLD remain poorly understood. We investigated the role and molecular mechanism of extracellular vesicles (EVs) released by hepatocytes during lipotoxicity in modulation of HSC phenotype. METHODS EVs were isolated from fat-laden hepatocytes by differential centrifugation and incubated with HSCs. EV internalization and HSCs activation, migration and proliferation were assessed. Loss- and gain-of-functions studies were performed to explore the potential role of PPAR-γ-targeting miRNAs carried by EVs into HSC. RESULTS Hepatocyte-derived EVs released during lipotoxicity are efficiently internalized by HSCs resulting in their activation, as shown by marked up-regulation of pro-fibrogenic genes (Collagen-I, α-SMA and TIMP-2), proliferation, chemotaxis and wound healing responses. These changes were associated with miRNAs shuttled by EVs and suppression of PPAR-γ expression in HSC. Hepatocyte-derived EVs miRNA content included various miRNAs that are known inhibitors of PPAR-γ expression with miR-128-3p being the most effectively transferred. Furthermore loss- and gain-of-function studies identified miR-128-3p as a central modulator of the effects of EVs on PPAR-γ inhibition and HSC activation. CONCLUSION Our findings demonstrate a link between fat-laden hepatocyte-derived EVs and liver fibrosis and have potential implications for the development of novel anti-fibrotic targets for NAFLD and other fibrotic diseases.
Collapse
Affiliation(s)
- Davide Povero
- Department of Pediatrics, University of California San Diego, La Jolla, California
| | - Nadia Panera
- Hepato-Metabolic Disease Unit and Liver Research Unit, Bambino-Gesu’ Children’s Hospital, Rome, Italy
| | - Akiko Eguchi
- Department of Pediatrics, University of California San Diego, La Jolla, California
| | - Casey D. Johnson
- Department of Pediatrics, University of California San Diego, La Jolla, California
| | | | - Lucas de Araujo Horcel
- Department of Pediatrics, University of California San Diego, La Jolla, California
- Centro Universitário Lusiada, Santos, Brazil
| | - Eva M. Pinatel
- Institute of Biomedical Technologies, National Research Council, Segrate, Italy
| | - Anna Alisi
- Hepato-Metabolic Disease Unit and Liver Research Unit, Bambino-Gesu’ Children’s Hospital, Rome, Italy
| | - Valerio Nobili
- Hepato-Metabolic Disease Unit and Liver Research Unit, Bambino-Gesu’ Children’s Hospital, Rome, Italy
| | - Ariel E. Feldstein
- Department of Pediatrics, University of California San Diego, La Jolla, California
- Correspondence Address correspondence to: Ariel E. Feldstein, MD, Division of Pediatric Gastroenterology, Hepatology, and Nutrition UCSD, 3020 Children’s Way, MC 5030, San Diego, California 92103–8450.
| |
Collapse
|
119
|
Tsukamoto H. Metabolic reprogramming and cell fate regulation in alcoholic liver disease. Pancreatology 2015; 15:S61-5. [PMID: 25800177 PMCID: PMC4515387 DOI: 10.1016/j.pan.2015.03.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 02/27/2015] [Accepted: 03/02/2015] [Indexed: 12/11/2022]
Abstract
UNLABELLED Alcoholic liver disease (ALD) should be defined as a life-style metabolic disease. Its pathogenesis is driven by altered cell fate of both parenchymal and non-parenchymal liver cell types, contributing to different pathologic spectra. A critical turning point in progression of ALD is chronic alcoholic steatohepatitis (ASH) or alcoholic neutrophilic hepatitis (AH), which markedly predisposes patients to most devastating ALD sequela, cirrhosis and liver cancer. RESULTS Our research identifies the pivotal roles of unique metabolic reprogramming in M1 activation of hepatic macrophages (HM) and myofibroblastic activation (MF) of hepatic stellate cells (HSC) in the genesis of inflammation and fibrosis, the two key histological features of chronic ASH and neutrophilic AH. For M1 HM activation, heightened proinflammatory iron redox signaling in endosomes or caveosomes results from altered iron metabolism and storage, promoting IKK/NF-kB activation via interactive activation of p21ras, TAK1, and PI3K. For MF cell fate regulation of HSC, activation of the morphogen Wnt pathway caused by the nuclear protein NECDIN or the single-pass trans-membrane protein DLK1, reprograms lipid metabolism via MeCP2-mediated epigenetic repression of the key HSC quiescence gene Ppar-γ. CONCLUSIONS The findings from these studies re-enforce the importance of metabolic reprogramming in cell fate regulation required for the pathogenesis of ALD.
Collapse
Affiliation(s)
- Hidekazu Tsukamoto
- Southern California Research Center ALPD and Cirrhosis and Department of Pathology, Keck School of Medicine of the University of Southern California, Greater Los Angeles VA Healthcare System, Los Angeles, California, USA
| |
Collapse
|
120
|
Fenofibrate is effective adjunctive therapy in the treatment of primary biliary cirrhosis: A meta-analysis. Clin Res Hepatol Gastroenterol 2015; 39:296-306. [PMID: 25882906 DOI: 10.1016/j.clinre.2015.02.011] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 02/24/2015] [Accepted: 02/27/2015] [Indexed: 02/04/2023]
Abstract
BACKGROUND AND AIM Fenofibrate is a potential novel therapy for primary biliary cirrhosis (PBC). We performed a systematic review and a meta-analysis of studies of fenofibrate in PBC. METHODS Electronic database search was performed for relevant studies. A search of abstracts presented in the main scientific meetings in the field and articles in press was also performed. Random effect model was used to pool the effect size across studies for changes in alkaline phosphatase, GGT, bilirubin and IgM levels before and after treatment and the overall rate of complete response to fenofibrate therapy. RESULTS Six studies with 102 patients met the inclusion criteria. All studies were case series and in all, patients who had no or incomplete response to UDCA had fenofibrate added at a dose of 100-200mg daily. Treatment duration ranged from 8-100weeks. Treatment with fenofibrate was associated with a significant decrease in alkaline phosphatase (-114IU/L, 95% CI: -152 to -76, P<0.0001); a significant decrease in GGT level (-92IU/L, 95% CI: -149 to -43; P=0.0004); significant decrease in total bilirubin (-0.11mg/dL, 95% CI: -0.18 to -0.08; P=0.0008); and a significant decrease in IgM level (-88mg/dL, 95% CI: -119 to -58; P<0.0001). The complete response rate was 69% (95% CI: 53-82%) with an odds ratio of 82.8 (95% CI: 21.6-317.2; P=0.024) while on fenofibrate. CONCLUSIONS Fenofibrate at doses of 100-200mg daily appears to be effective adjunctive therapy in PBC patients who had no or incomplete response to UDCA. There is a critical need for larger scale randomized trials to determine its effect on liver-related morbidity and mortality (or progression towards end-stage disease).
Collapse
|
121
|
Tang Y. Curcumin targets multiple pathways to halt hepatic stellate cell activation: updated mechanisms in vitro and in vivo. Dig Dis Sci 2015; 60:1554-64. [PMID: 25532502 DOI: 10.1007/s10620-014-3487-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 12/07/2014] [Indexed: 12/12/2022]
Abstract
Nonalcoholic steatohepatitis (NASH) is the advanced form of nonalcoholic fatty liver disease, which is often accompanied by obese and/or type II diabetes mellitus. Approximately one-third of NASH patients develop hepatic fibrosis. Hepatic stellate cells are the major effector cells during liver fibrogenesis. Advanced liver fibrosis usually proceeds to cirrhosis and even hepatocellular carcinoma, leading to liver failure, portal hypertension and even death. Currently, there are no approved agents for treatment and prevention of liver fibrosis in human beings. Curcumin, the principal curcuminoid of turmeric, has been reported to show antitumor, antioxidant, and anti-inflammatory properties both in in vitro and in vivo systems. Accumulating data shows that curcumin plays a critical role in combating liver fibrogenesis. This review will discuss the inhibitory roles of curcumin and update the underlying mechanisms by which curcumin targets in inhibiting hepatic stellate cell activation.
Collapse
Affiliation(s)
- Youcai Tang
- Department of Pediatrics, The Second Affiliated Hospital, Zhengzhou University, 2 Jingba Road, Zhengzhou, 450014, Henan, China,
| |
Collapse
|
122
|
Jiang Y, Wang S, Zhao Y, Lin C, Zhong F, Jin L, He F, Wang H. Histone H3K9 demethylase JMJD1A modulates hepatic stellate cells activation and liver fibrosis by epigenetically regulating peroxisome proliferator‐activated receptor γ. FASEB J 2015; 29:1830-1841. [DOI: 10.1096/fj.14-251751] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Affiliation(s)
- Yan Jiang
- State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life SciencesShanghaiChina
- Institutes of Biomedical Sciences of Shanghai Medical College, Fudan UniversityShanghaiChina
- Department of ChemistryFudan UniversityShanghaiChina
| | - Sheng Wang
- State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life SciencesShanghaiChina
- Institutes of Biomedical Sciences of Shanghai Medical College, Fudan UniversityShanghaiChina
| | - Yuanyuan Zhao
- State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life SciencesShanghaiChina
- Institutes of Biomedical Sciences of Shanghai Medical College, Fudan UniversityShanghaiChina
| | - Chengzhao Lin
- State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life SciencesShanghaiChina
- Institutes of Biomedical Sciences of Shanghai Medical College, Fudan UniversityShanghaiChina
| | - Fan Zhong
- State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life SciencesShanghaiChina
- Institutes of Biomedical Sciences of Shanghai Medical College, Fudan UniversityShanghaiChina
| | - Li Jin
- State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life SciencesShanghaiChina
- Institutes of Biomedical Sciences of Shanghai Medical College, Fudan UniversityShanghaiChina
| | - Fuchu He
- State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life SciencesShanghaiChina
- Institutes of Biomedical Sciences of Shanghai Medical College, Fudan UniversityShanghaiChina
- Department of ChemistryFudan UniversityShanghaiChina
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation MedicineBeijingChina
| | - Haijian Wang
- State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life SciencesShanghaiChina
- Institutes of Biomedical Sciences of Shanghai Medical College, Fudan UniversityShanghaiChina
| |
Collapse
|
123
|
Abstract
PURPOSE OF REVIEW Nonalcoholic fatty liver disease is the most common cause of liver dysfunction in the western world because of its close association with obesity, insulin resistance and dyslipidaemia. Nonalcoholic steatohepatitis (NASH) is a particular health concern due to the increased morbidity and mortality associated with progressive disease. At present, without specific targeted pharmacological therapies, the mainstay of therapy remains weight loss through dietary modification and lifestyle change; thus, the purpose of this review is to summarize the recent evidence for current and emerging therapies in NASH. RECENT FINDINGS Some existing medications, including pioglitazones and angiotensin receptor antagonists, may be repurposed to help treat this condition. Vitamin E may improve histology in NASH, but safety issues limit its use. Recently, a number of novel agents specifically targeting nonalcoholic fatty liver disease pathogenesis have entered clinical trials, including the farnesoid X receptor agonist obeticholic acid, which has shown significant histological improvements in steatohepatitis and fibrosis. SUMMARY Diet/lifestyle modification remains the mainstay of treatment. For patients with NASH and advanced fibrosis, current liver-directed pharmacotherapy with vitamin E and pioglitazone offer some benefits; obeticholic acid appears promising and is currently being tested. Comorbidities must be diagnosed and treated; cardiovascular disease remains a primary cause of death in these patients.
Collapse
|
124
|
Ramani K, Donoyan S, Tomasi ML, Park S. Role of methionine adenosyltransferase α2 and β phosphorylation and stabilization in human hepatic stellate cell trans-differentiation. J Cell Physiol 2015; 230:1075-85. [PMID: 25294683 DOI: 10.1002/jcp.24839] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 09/22/2014] [Indexed: 12/14/2022]
Abstract
Myofibroblastic trans-differentiation of hepatic stellate cells (HSCs) is an essential event in the development of liver fibrogenesis. These changes involve modulation of key regulators of the genome and the proteome. Methionine adenosyltransferases (MAT) catalyze the biosynthesis of the methyl donor, S-adenosylmethionine (SAMe) from methionine. We have previously shown that two MAT genes, MAT2A and MAT2B (encoding MATα2 and MATβ proteins respectively), are required for HSC activation and loss of MAT2A transcriptional control favors its up-regulation during trans-differentiation. Hence MAT genes are intrinsically linked to the HSC machinery during activation. In the current study, we have identified for the first time, post-translational modifications in the MATα2 and MATβ proteins that stabilize them and favor human HSC trans-differentiation. Culture-activation of human HSCs induced the MATα2 and MATβ proteins. Using mass spectrometry, we identified phosphorylation sites in MATα2 and MATβ predicted to be phosphorylated by mitogen-activated protein kinase (MAPK) family members (ERK1/2, V-Raf Murine Sarcoma Viral Oncogene Homolog B1 [B-Raf], MEK). Phosphorylation of both proteins was enhanced during HSC activation. Blocking MEK activation lowered the phosphorylation and stability of MAT proteins without influencing their mRNA levels. Silencing ERK1/2 or B-Raf lowered the phosphorylation and stability of MATβ but not MATα2. Reversal of the activated human HSC cell line, LX2 to quiescence lowered phosphorylation and destabilized MAT proteins. Mutagenesis of MATα2 and MATβ phospho-sites destabilized them and prevented HSC trans-differentiation. The data reveal that phosphorylation of MAT proteins during HSC activation stabilizes them thereby positively regulating trans-differentiation.
Collapse
Affiliation(s)
- Komal Ramani
- Division of Gastroenterology and Liver Diseases, USC Research Center for Liver Diseases, Keck School of Medicine University of Southern California, Los Angeles, California
| | | | | | | |
Collapse
|
125
|
Hou G, Yin Y, Han D, Wang QY, Kang J. Rosiglitazone attenuates the metalloprotease/anti-metalloprotease imbalance in emphysema induced by cigarette smoke: involvement of extracellular signal-regulated kinase and NFκB signaling. Int J Chron Obstruct Pulmon Dis 2015; 10:715-24. [PMID: 25897215 PMCID: PMC4396520 DOI: 10.2147/copd.s77514] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Objective We investigated how rosiglitazone attenuated cigarette smoke (CS)-induced emphysema in a rat model. In particular, we focused on its possible effects on the imbalance between metalloprotease (MMP) and anti-MMP activity, mitogen-activated protein kinase (MAPK) phosphorylation, and nuclear factor kappa-light-chain-enhancer of activated B cell (NFκB) signaling pathway over-activation. Methods A total of 36 Wistar rats were divided into three groups (n=12 each): animals were exposed to CS for 12 weeks in the absence (the CS group) or presence of 30 mg/kg rosiglitazone (the rosiglitazone-CS [RCS] group); a control group was treated with the rosiglitazone vehicle only, without any CS exposure. Histopathology of lung tissue in all groups was evaluated to grade severity of the disease. Expression levels of peroxisome proliferator-activated receptor γ (PPARγ), MMP2, and MMP9 in lung tissue were determined and compared using Western blotting and immunohistochemistry. Activation of MAPKs, NFκB, and the nuclear factor of kappa light polypeptide gene enhancer in B-cell inhibitor, alpha (IκBα) phosphorylation in lung tissue was examined by Western blotting. Results Emphysema-related pathology, based on inter-alveolar wall distance and alveolar density, was less severe in the RCS group than in the CS group. Compared with the CS group, levels of PPARγ were higher in the RCS group, and levels of MMP2 and MMP9 proteins were lower in the RCS rats. Levels of activated MAPKs and NFκB were also lower, while the IκBαphosphorylation was increased in the lung tissue of RCS rats. Conclusion Our findings suggest that oral administration of rosiglitazone attenuates the metalloprotease activity induced by CS, and the underlying mechanism might involve the activation of signaling pathways dependent on MAPKs or NFκB. Our results further suggest that PPARγ contributes to the pathogenesis of emphysema as well as airway inflammation induced by CS.
Collapse
Affiliation(s)
- Gang Hou
- Department of Respiratory Medicine, the First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Yan Yin
- Department of Respiratory Medicine, the First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Dan Han
- Department of Respiratory Medicine, the First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Qiu-Yue Wang
- Department of Respiratory Medicine, the First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Jian Kang
- Department of Respiratory Medicine, the First Hospital of China Medical University, Shenyang, People's Republic of China
| |
Collapse
|
126
|
Lee JW, Asai M, Jeon SK, Iimura T, Yonezawa T, Cha BY, Woo JT, Yamaguchi A. Rosmarinic acid exerts an antiosteoporotic effect in the RANKL-induced mouse model of bone loss by promotion of osteoblastic differentiation and inhibition of osteoclastic differentiation. Mol Nutr Food Res 2015; 59:386-400. [PMID: 25380345 DOI: 10.1002/mnfr.201400164] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Revised: 10/17/2014] [Accepted: 10/20/2014] [Indexed: 12/25/2022]
Abstract
SCOPE Bone homeostasis is ensured by the balance between bone formation and resorption. Thus, control of the recruitment, proliferation, and differentiation of bone cells is essential to maintain bone mass. The aim of this study was to elucidate the effects of rosmarinic acid as a potential therapeutic agent on bone metabolism using bone cells and a mouse model. METHODS AND RESULTS Rosmarinic acid increased alkaline phosphatase activity and induced mineralization in osteoblasts. Addition of rosmarinic acid to cultures of calvarial osteoblastic cells prepared from T-cell factor/β-catenin TOP-GAL mutant mice strongly induced the expression of LacZ and promoted stabilization of β-catenin in the cytoplasm of ST2 cells, suggesting that rosmarinic acid affects the canonical Wnt signaling pathway. Moreover, rosmarinic acid inhibited not only osteoclast formation in cocultures of mouse bone marrow cells and osteoblasts, but also receptor activator of nuclear factor kappa-B ligand (RANKL)-induced osteoclastic differentiation in bone marrow-derived macrophages. RANKL-induced p38 mitogen-activated protein kinase and the expression of nuclear factor of activated T cell, c-Jun, and c-Fos were inhibited by rosmarinic acid in bone marrow macrophages. Finally, we confirmed that rosmarinic acid improved bone mass in a soluble RANKL-induced bone loss mouse model. CONCLUSION Rosmarinic acid has dual regulatory effects on bone metabolism and may control the bone functions by controlling osteoblastic and osteoclastic differentiation.
Collapse
Affiliation(s)
- Ji-Won Lee
- Division of Bio-Imaging, Proteo-Science Center (PROS), Ehime University, Ehime, Japan; Section of Oral Pathology, Graduate School of Medicine and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Research Institute for Biological Functions, Chubu University, Aichi, Japan
| | | | | | | | | | | | | | | |
Collapse
|
127
|
Hayashizaki-Someya Y, Kurosaki E, Takasu T, Mitori H, Yamazaki S, Koide K, Takakura S. Ipragliflozin, an SGLT2 inhibitor, exhibits a prophylactic effect on hepatic steatosis and fibrosis induced by choline-deficient l-amino acid-defined diet in rats. Eur J Pharmacol 2015; 754:19-24. [PMID: 25701721 DOI: 10.1016/j.ejphar.2015.02.009] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Revised: 02/03/2015] [Accepted: 02/10/2015] [Indexed: 12/11/2022]
Abstract
Ipragliflozin is a selective sodium glucose cotransporter 2 (SGLT2) inhibitor that increases urinary glucose excretion by inhibiting renal glucose reabsorption and thereby causes a subsequent antihyperglycemic effect. As nonalcoholic fatty liver disease (NAFLD), including nonalcoholic steatohepatitis (NASH), is closely linked to metabolic diseases such as obesity and diabetes, we investigated the effect of ipragliflozin on NAFLD in rats fed a choline-deficient l-amino acid-defined (CDAA) diet. Five weeks after starting the CDAA diet, rats exhibited hepatic triglyceride (TG) accumulation, fibrosis, and mild inflammation. Repeated oral administration of ipragliflozin (3mg/g, once daily for 5 weeks) prevented both hepatic TG accumulation (188 vs.290 mg/g tissue vehicle-treated group; P<0.001) and large lipid droplet formation. Further, ipragliflozin exerted a prophylactic effect on liver fibrosis, as indicated by a marked decrease in hydroxyproline content and fibrosis score. Pioglitazone, which is known to be effective on hepatic fibrosis in CDAA diet-fed rats as well as NASH patients with type 2 diabetes mellitus (T2DM), also exerted a mild prophylactic effect on fibrosis, but not on hepatic TG accumulation or inflammation. In conclusion, ipragliflozin prevented hepatic TG accumulation and fibrosis in CDAA-diet rats. These findings suggest the therapeutic potential of ipragliflozin for patients with NAFLD.
Collapse
Affiliation(s)
| | - Eiji Kurosaki
- Drug Discovery Research, Astellas Pharma Inc., Tsukuba, Ibaraki 305-8585, Japan
| | - Toshiyuki Takasu
- Drug Discovery Research, Astellas Pharma Inc., Tsukuba, Ibaraki 305-8585, Japan
| | - Hikaru Mitori
- Drug Discovery Research, Astellas Pharma Inc., Tsukuba, Ibaraki 305-8585, Japan
| | - Shunji Yamazaki
- Drug Discovery Research, Astellas Pharma Inc., Tsukuba, Ibaraki 305-8585, Japan
| | - Kumi Koide
- Drug Discovery Research, Astellas Pharma Inc., Tsukuba, Ibaraki 305-8585, Japan
| | - Shoji Takakura
- Drug Discovery Research, Astellas Pharma Inc., Tsukuba, Ibaraki 305-8585, Japan
| |
Collapse
|
128
|
Nair DG, Weiskirchen R, Al-Musharafi SK. The use of marine-derived bioactive compounds as potential hepatoprotective agents. Acta Pharmacol Sin 2015; 36:158-70. [PMID: 25500871 DOI: 10.1038/aps.2014.114] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Accepted: 09/26/2014] [Indexed: 12/20/2022] Open
Abstract
The marine environment may be explored as a rich source for novel drugs. A number of marine-derived compounds have been isolated and identified, and their therapeutic effects and pharmacological profiles are characterized. In the present review, we highlight the recent studies using marine compounds as potential hepatoprotective agents for the treatment of liver fibrotic diseases and discuss the proposed mechanisms of their activities. In addition, we discuss the significance of similar studies in Oman, where the rich marine life provides a potential for the isolation of novel natural, bioactive products that display therapeutic effects on liver diseases.
Collapse
|
129
|
Tuohetahuntila M, Spee B, Kruitwagen HS, Wubbolts R, Brouwers JF, van de Lest CH, Molenaar MR, Houweling M, Helms JB, Vaandrager AB. Role of long-chain acyl-CoA synthetase 4 in formation of polyunsaturated lipid species in hepatic stellate cells. Biochim Biophys Acta Mol Cell Biol Lipids 2014; 1851:220-30. [PMID: 25500141 DOI: 10.1016/j.bbalip.2014.12.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Revised: 11/17/2014] [Accepted: 12/01/2014] [Indexed: 02/02/2023]
Abstract
Hepatic stellate cell (HSC) activation is a critical step in the development of chronic liver disease. We previously observed that the levels of triacylglycerol (TAG) species containing long polyunsaturated fatty acids (PUFAs) are increased in in vitro activated HSCs. Here we investigated the cause and consequences of the rise in PUFA-TAGs by profiling enzymes involved in PUFA incorporation. We report that acyl CoA synthetase (ACSL) type 4, which has a preference for PUFAs, is the only upregulated ACSL family member in activated HSCs. Inhibition of the activity of ACSL4 by siRNA-mediated knockdown or addition of rosiglitazone specifically inhibited the incorporation of deuterated arachidonic acid (AA-d8) into TAG in HSCs. In agreement with this, ACSL4 was found to be partially localized around lipid droplets (LDs) in HSCs. Inhibition of ACSL4 also prevented the large increase in PUFA-TAGs in HSCs upon activation and to a lesser extent the increase of arachidonate-containing phosphatidylcholine species. Inhibition of ACSL4 by rosiglitazone was associated with an inhibition of HSC activation and prostaglandin secretion. Our combined data show that upregulation of ACSL4 is responsible for the increase in PUFA-TAG species during activation of HSCs, which may serve to protect cells against a shortage of PUFAs required for eicosanoid secretion.
Collapse
Affiliation(s)
- Maidina Tuohetahuntila
- Department of Biochemistry and Cell Biology, Faculty of Veterinary Medicine & Institute of Biomembranes, Utrecht University, Yalelaan 2, 3584 CM Utrecht, The Netherlands.
| | - Bart Spee
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 104, 3584 CM Utrecht, The Netherlands.
| | - Hedwig S Kruitwagen
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 104, 3584 CM Utrecht, The Netherlands.
| | - Richard Wubbolts
- Department of Biochemistry and Cell Biology, Faculty of Veterinary Medicine & Institute of Biomembranes, Utrecht University, Yalelaan 2, 3584 CM Utrecht, The Netherlands.
| | - Jos F Brouwers
- Department of Biochemistry and Cell Biology, Faculty of Veterinary Medicine & Institute of Biomembranes, Utrecht University, Yalelaan 2, 3584 CM Utrecht, The Netherlands.
| | - Chris H van de Lest
- Department of Biochemistry and Cell Biology, Faculty of Veterinary Medicine & Institute of Biomembranes, Utrecht University, Yalelaan 2, 3584 CM Utrecht, The Netherlands.
| | - Martijn R Molenaar
- Department of Biochemistry and Cell Biology, Faculty of Veterinary Medicine & Institute of Biomembranes, Utrecht University, Yalelaan 2, 3584 CM Utrecht, The Netherlands.
| | - Martin Houweling
- Department of Biochemistry and Cell Biology, Faculty of Veterinary Medicine & Institute of Biomembranes, Utrecht University, Yalelaan 2, 3584 CM Utrecht, The Netherlands.
| | - J Bernd Helms
- Department of Biochemistry and Cell Biology, Faculty of Veterinary Medicine & Institute of Biomembranes, Utrecht University, Yalelaan 2, 3584 CM Utrecht, The Netherlands.
| | - Arie B Vaandrager
- Department of Biochemistry and Cell Biology, Faculty of Veterinary Medicine & Institute of Biomembranes, Utrecht University, Yalelaan 2, 3584 CM Utrecht, The Netherlands.
| |
Collapse
|
130
|
Zhao XR, Gonzales N, Aronowski J. Pleiotropic role of PPARγ in intracerebral hemorrhage: an intricate system involving Nrf2, RXR, and NF-κB. CNS Neurosci Ther 2014; 21:357-66. [PMID: 25430543 DOI: 10.1111/cns.12350] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Revised: 10/10/2014] [Accepted: 10/11/2014] [Indexed: 12/13/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is a subtype of stroke involving formation of hematoma within brain parenchyma, which accounts for 8-15% of all strokes in Western societies and 20-30% among Asian populations, and has a 1-year mortality rate >50%. The high mortality and severe morbidity make ICH a major public health problem. Only a few evidence-based targeted treatments are used for ICH management, and interventions focus primarily on supportive care and comorbidity prevention. Even in patients who survive the ictus, extravasated blood (including plasma components) and subsequent intrahematoma hemolytic products trigger a series of adverse events within the brain parenchyma, leading to secondary brain injury, edema and severe neurological deficits or death. Although the hematoma in humans gradually resolves within months, full restoration of neurological function can be slow and often incomplete, leaving survivors with devastating neurological deficits. During past years, peroxisome proliferator-activated receptor gamma (PPARγ) transcription factor and its agonists received recognition as important players in regulating not only glucose and lipid metabolism (which underlies its therapeutic effect in type 2 diabetes mellitus), and more recently, as an instrumental pleiotropic regulator of antiinflammation, antioxidative regulation, and phagocyte-mediated cleanup processes. PPARγ agonists have emerged as potential therapeutic target for stroke. The use of PPARγ as a therapeutic target appears to have particularly strong compatibility toward pathogenic components of ICH. In addition to its direct genomic effect, PPARγ may interact with transcription factor, NF-κB, which may underlie many aspects of the antiinflammatory effect of PPARγ. Furthermore, PPARγ appears to regulate expression of Nrf2, another transcription factor and master regulator of detoxification and antioxidative regulation. Finally, the synergistic costimulation of PPARγ and retinoid X receptor, RXR, may play an additional role in the therapeutic modulation of PPARγ function. In this article, we outline the main components of the role of PPARγ in ICH pathogenesis.
Collapse
Affiliation(s)
- Xiu-Rong Zhao
- Department of Neurology, Stroke Research Center, University of Texas Medical School - Houston, Houston, TX, USA
| | | | | |
Collapse
|
131
|
Zhou Q, Guan W, Qiao H, Cheng Y, Li Z, Zhai X, Zhou Y. GATA binding protein 2 mediates leptin inhibition of PPARγ1 expression in hepatic stellate cells and contributes to hepatic stellate cell activation. Biochim Biophys Acta Mol Basis Dis 2014; 1842:2367-77. [PMID: 25305367 DOI: 10.1016/j.bbadis.2014.10.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2014] [Revised: 09/15/2014] [Accepted: 10/01/2014] [Indexed: 02/06/2023]
Abstract
Hepatic stellate cell (HSC) activation is a crucial step in the development of liver fibrosis. Peroxisome-proliferator activated receptor γ (PPARγ) exerts a key role in the inhibition of HSC activation. Leptin reduces PPARγ expression in HSCs and plays a unique role in promoting liver fibrosis. The present studies aimed to investigate the mechanisms underlying leptin regulation of PPARγ1 (a major subtype of PPARγ) in HSCs in vivo and in vitro. Results revealed a leptin response region in mouse PPARγ1 promoter and indicated that the region included a GATA binding protein binding site around position -2323. GATA binding protein-2 (GATA-2) could bind to the site and inhibit PPARγ1 promoter activity in HSCs. Leptin induced GATA-2 expression in HSCs in vitro and in vivo. GATA-2 mediated leptin inhibition of PPARγ1 expression by its binding site in PPARγ1 promoter in HSCs and GATA-2 promoted HSC activation. Leptin upregulated GATA-2 expression through β-catenin and sonic hedgehog pathways in HSCs. Leptin-induced increase in GATA-2 was accompanied by the decrease in PPARγ expression in HSCs and by the increase in the activated HSC number and liver fibrosis in vivo. Our data might suggest a possible new explanation for the promotion effect of leptin on liver fibrogenesis.
Collapse
Affiliation(s)
- Qian Zhou
- Department of Pharmacology, School of Pharmacy, Nantong University, Qi Xiou Road 19, Nantong 226001, Jiangsu, China
| | - Wei Guan
- Department of Pharmacology, School of Pharmacy, Nantong University, Qi Xiou Road 19, Nantong 226001, Jiangsu, China
| | - Haowen Qiao
- Department of Biochemistry & Molecular Biology, Medical College, Nantong University, Qi Xiou Road 19, Nantong, 226001, Jiangsu, China
| | - Yuanyuan Cheng
- Department of Pharmacology, School of Pharmacy, Nantong University, Qi Xiou Road 19, Nantong 226001, Jiangsu, China
| | - Ziqiang Li
- Department of Biochemistry & Molecular Biology, Medical College, Nantong University, Qi Xiou Road 19, Nantong, 226001, Jiangsu, China
| | - Xuguang Zhai
- Department of Biochemistry & Molecular Biology, Medical College, Nantong University, Qi Xiou Road 19, Nantong, 226001, Jiangsu, China
| | - Yajun Zhou
- Department of Biochemistry & Molecular Biology, Medical College, Nantong University, Qi Xiou Road 19, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
132
|
Cho YM, Kwak SN, Joo NS, Kim DH, Lee AH, Kim KS, Seo JB, Jeong SW, Kwon OJ. X-box binding protein 1 is a novel key regulator of peroxisome proliferator-activated receptor γ2. FEBS J 2014; 281:5132-46. [PMID: 25223794 DOI: 10.1111/febs.13052] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 08/10/2014] [Accepted: 09/12/2014] [Indexed: 01/08/2023]
Abstract
X-box binding protein 1 (XBP1), a transcription factor of the unfolded protein response, plays various roles in many biological processes. We examined its pro-adipogenic activity and target genes during adipogenic differentiation in wild-type and genetically modified 3T3-L1 cells. Signalling pathways that contribute to Xbp1 mRNA splicing, and the correlation of the transcriptionally active XBP1 isoform (XBP1s) level with body mass index and the level of peroxisome proliferator-activated receptor γ2 (PPARγ2) in human adipose tissues were also examined. The mRNA and nuclear protein expression levels of XBP1s increased immediately following hormonal induction of adipogenesis, reaching a peak at 6 h. Results from cDNA microarray and gene expression analyses using genetically modified cells indicated that PPARγ2 was a principal target of XBP1s. The XBP1s-specific binding motif, which is distinct from the CCAAT/enhancer-binding protein α binding site, was identified in the PPARγ2 promoter by site-directed mutagenesis. Fetal bovine serum, insulin, 3-isobutyl-1-methylxanthine and dexamethasone contributed independently to Xbp1 mRNA splicing. In human subcutaneous adipose tissues, the levels of both Xbp1s and Pparγ2 mRNA increased proportionally with body mass index, and there was a significant positive correlation between the two genes. These data suggest for the first time that positive regulation of PPARγ2 is a principal mechanism of XBP1s-mediated adipogenesis in 3T3-L1 cells.
Collapse
Affiliation(s)
- Yoon Mi Cho
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
133
|
Mahmoud AM. Hesperidin protects against cyclophosphamide-induced hepatotoxicity by upregulation of PPARγ and abrogation of oxidative stress and inflammation. Can J Physiol Pharmacol 2014; 92:717-24. [DOI: 10.1139/cjpp-2014-0204] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The most important reason for the non-approval and withdrawal of drugs by the Food and Drug Administration is hepatotoxicity. Therefore, this study was undertaken to evaluate the protective effects of hesperidin against cyclophosphamide (CYP)-induced hepatotoxicity in Wistar rats. The rats received a single intraperitoneal dose of CYP of 200 mg/kg body mass, followed by treatment with hesperidin, orally, at doses of 25 and 50 mg/kg for 11 consecutive days. CYP induced hepatic damage, as evidenced by the significantly elevated levels of serum pro-inflammatory cytokines, serum transaminases, liver lipid peroxidation, and nitric oxide. As a consequence, there was reduced glutathione content, and the activities of the antioxidant enzymes superoxide dismutase, catalase, and glutathione peroxidase, were markedly reduced. In addition, CYP administration induced a considerable downregulation of peroxisome proliferator activated receptor gamma (PPARγ) and upregulation of nuclear factor-kappa B (NF-κB) and inducible nitric oxide synthase (iNOS) mRNA expression. Hesperidin, in a dose-dependent manner, rejuvenated the altered markers to an almost normal state. In conclusion, hesperidin showed a potent protective effect against CYP-induced oxidative stress and inflammation leading to hepatotoxicity. The study suggests that hesperidin exerts its protective effect against CYP-induced hepatotoxicity through upregulation of hepatic PPARγ expression and abrogation of inflammation and oxidative stress.
Collapse
Affiliation(s)
- Ayman M. Mahmoud
- Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, 62514 Beni-Suef, Egypt
| |
Collapse
|
134
|
Liu Q, Wang CY, Liu Z, Ma XS, He YH, Chen SS, Bai XY. Hydroxysafflor yellow A suppresses liver fibrosis induced by carbon tetrachloride with high-fat diet by regulating PPAR-γ/p38 MAPK signaling. PHARMACEUTICAL BIOLOGY 2014; 52:1085-1093. [PMID: 24618007 DOI: 10.3109/13880209.2013.877491] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
CONTEXT One approach to protect against liver fibrosis is the use of herb-derived natural compounds, such as hydroxysafflor yellow A (HSYA). The antifibrosis effect of HYSA against liver fibrosis has been investigated; however, its mechanisms have not yet been entirely revealed. OBJECTIVES To study the protective effects of HSYA on liver fibrosis induced by carbon tetrachloride (CCl4) and a high-fat diet (HFD), and to determine the mechanism of action of HSYA. MATERIALS AND METHODS CCl4 and HFD were used to mimic liver fibrosis in rats, and serum biochemical indicators were determined. The antifibrosis effects of HSYA were evaluated and its mechanisms were investigated by histopathological analysis, immunohistochemical staining, enzyme-linked immunosorbent assays, real-time-PCR, and western blotting. RESULTS HSYA reduced CCl4- and HFD-mediated liver fibrosis and ameliorated serum biochemical indicator, downregulated the expression of tissue inhibitor of metalloproteinase-1 (TIMP-1) (0.31 ± 0.03 protein, 0.59 ± 0.02 mRNA) and transformin growth factor-β1 (TGF-β1) (0.81 ± 0.02 protein, 0.58 ± 0.04 mRNA), and upregulated the expression of peroxisome proliferator-activated receptor-γ (PPAR-γ) (1.57 ± 0.13 protein, 2.48 ± 0.19 mRNA) and matrix metallopeptidases-2 (MMP-2) (2.31 ± 0.16 protein, 2.79 ± 0.22 mRNA) (p < 0.01, versus model group). These effects were significantly attenuated by PPAR-γ antagonist GW9662 via blocking the phosphorylation of p38 MAPK. DISCUSSION AND CONCLUSION These data demonstrate a novel role for HSYA in inhibiting CCl4- and HFD-mediated liver fibrosis, and reveal that PPAR-γ and p38 MAPK signaling play pivotal roles in the prevention of liver fibrosis induced by CCl4 and HFD.
Collapse
Affiliation(s)
- Q Liu
- School of Pharmaceutical Sciences, Binzhou Medical University , Yantai, Shandong , PR China
| | | | | | | | | | | | | |
Collapse
|
135
|
Lee KW, Thiyagarajan V, Sie HW, Cheng MF, Tsai MJ, Chia YC, Weng CF. Synergistic effect of natural compounds on the fatty acid-induced autophagy of activated hepatic stellate cells. J Nutr Biochem 2014; 25:903-13. [DOI: 10.1016/j.jnutbio.2014.04.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 03/23/2014] [Accepted: 04/06/2014] [Indexed: 01/22/2023]
|
136
|
A PPARγ, NF-κB and AMPK-dependent mechanism may be involved in the beneficial effects of curcumin in the diabetic db/db mice liver. Molecules 2014; 19:8289-302. [PMID: 24945581 PMCID: PMC6271620 DOI: 10.3390/molecules19068289] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Revised: 06/06/2014] [Accepted: 06/12/2014] [Indexed: 02/06/2023] Open
Abstract
Turmeric (Curcuma longa) is a rhizomatous herbaceous perennial plant of the ginger family which has been used to treat biliary disorders, anorexia, cough, rheumatism, cancer, sinusitis, hepatic disorders, hyperglycemia, obesity, and diabetes in both Ayurvedic and Traditional Chinese Medicine. Suggested mechanisms of action include the modulation of signal transduction cascades and effects on gene expression, however they remain to be elucidated. In this study, the expression of some proteins responsible for transcription factors, inflammation, and metabolic control were evaluated by western blot in 15-week-old db/db mice livers treated with curcumin 0.75% mixed in their diet for 8 weeks. In addition, nitrosative stress was evaluated. Curcumin increased the expression of AMPK and PPARγ, and diminished NF-κB protein in db/db mice. However, it did not modify the expression of PGC-1α or SIRT1. Nitrosative stress present in db/db mice livers was determined by a unique nitrotyrosylated protein band (75 kDa) and was not reverted with curcumin. In conclusion, curcumin regulates the expression of AMPK, PPARγ, and NF-κB; suggesting a beneficial effect for treatment of T2DM complications. In order to observe best beneficial effects it is desirable to administer curcumin in the earlier states of T2DM.
Collapse
|
137
|
Tsolaki E, Athanasiou E, Gounari E, Zogas N, Siotou E, Yiangou M, Anagnostopoulos A, Yannaki E. Hematopoietic stem cells and liver regeneration: differentially acting hematopoietic stem cell mobilization agents reverse induced chronic liver injury. Blood Cells Mol Dis 2014; 53:124-32. [PMID: 24923531 DOI: 10.1016/j.bcmd.2014.05.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Revised: 02/02/2014] [Accepted: 02/03/2014] [Indexed: 12/20/2022]
Abstract
Bone marrow (BM) could serve as a source of cells facilitating liver repopulation in case of hepatic damage. Currently available hematopoietic stem cell (HSC) mobilizing agents, were comparatively tested for healing potential in liver fibrosis. Carbon tetrachloride (CCl4)-injured mice previously reconstituted with Green Fluorescent Protein BM were mobilized with Granulocyte-Colony Stimulating Factor (G-CSF), Plerixafor or G-CSF+Plerixafor. Hepatic fibrosis, stellate cell activation and oval stem cell frequency were measured by Gomori and by immunohistochemistry for a-Smooth Muscle Actin and Cytokeratin-19, respectively. Angiogenesis was evaluated by ELISA and immunohistochemistry. Quantitative real-time PCR was used to determine the mRNA levels of liver Peroxisome Proliferator-Activated Receptor gamma (PPAR-γ), Interleukin-6 (IL-6) and Tumor Necrosis-alpha (TNFα). BM-derived cells were tracked by double immunofluorescence. The spontaneous migration of mobilized HSCs towards injured liver and its cytokine secretion profile was determined in transwell culture systems. Either single-agent mobilization or the combination of agents significantly ameliorated hepatic damage by decreasing fibrosis and restoring the abnormal vascular network in the liver of mobilized mice compared to CCl4-only mice. The degree of fibrosis reduction was similar among all mobilized mice despite that G-CSF+Plerixafor yielded significantly higher numbers of circulating HSCs over other agents. The liver homing potential of variously mobilized HSCs differed among the agents. An extended G-CSF treatment provided the highest anti-fibrotic effect over all tested modalities, induced by the proliferation of hepatic stem cells and decreased hepatic inflammation. Plerixafor-mobilized HSCs, despite their reduced liver homing potential, reversed fibrosis mainly by increasing hepatic PPAR-γ and VEGF expression. In all groups, BM-derived mature hepatocytes as well as liver-committed BM stem cells were detected only at low frequencies, further supporting the concept that alternative mechanisms rather than direct HSC effects regulate liver recovery. Overall, our data suggest that G-CSF, Plerixafor and G-CSF+Plerixafor act differentially during the wound healing process, ultimately providing a potent anti-fibrotic effect.
Collapse
Affiliation(s)
- Eleftheria Tsolaki
- Gene and Cell Therapy Center, Hematology Department-BMT Unit, George Papanicolaou Hospital, Thessaloniki, Greece; Department of Genetics, Development and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Evangelia Athanasiou
- Gene and Cell Therapy Center, Hematology Department-BMT Unit, George Papanicolaou Hospital, Thessaloniki, Greece
| | - Eleni Gounari
- Gene and Cell Therapy Center, Hematology Department-BMT Unit, George Papanicolaou Hospital, Thessaloniki, Greece; Department of Genetics, Development and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Nikolaos Zogas
- Gene and Cell Therapy Center, Hematology Department-BMT Unit, George Papanicolaou Hospital, Thessaloniki, Greece; Department of Genetics, Development and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Eleni Siotou
- Gene and Cell Therapy Center, Hematology Department-BMT Unit, George Papanicolaou Hospital, Thessaloniki, Greece
| | - Minas Yiangou
- Department of Genetics, Development and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Achilles Anagnostopoulos
- Gene and Cell Therapy Center, Hematology Department-BMT Unit, George Papanicolaou Hospital, Thessaloniki, Greece
| | - Evangelia Yannaki
- Gene and Cell Therapy Center, Hematology Department-BMT Unit, George Papanicolaou Hospital, Thessaloniki, Greece.
| |
Collapse
|
138
|
Abstract
PURPOSE OF REVIEW Primary biliary cirrhosis (PBC) can lead to end-stage liver disease and death. Ursodeoxycholic acid (UDCA) treatment can normalize serum liver enzymes in PBC, and such UDCA-responsive patients have a similar life expectancy as age and sex-matched controls. Nearly up to 50% of the patients with PBC, depending on sex and age at diagnosis, show an incomplete biochemical response to UDCA and require additional/alternative treatment. The purpose of this review is to critically evaluate the molecular mechanisms and clinical benefit of fibrate treatment in these patients. RECENT FINDINGS Fibrates have anticholestatic, anti-inflammatory, and antifibrotic effects in animal and in-vitro studies. The mechanisms that underlie these effects are complementary, and largely mediated through activation of peroxisome proliferator activated receptors. Fibrate treatment ameliorated liver biochemical tests in UDCA unresponsive patients, either as mono-therapy or in combination with UDCA. These results, however, were obtained in case series and small pilot studies. The results of phase III studies, such as the Bezafibrate in Combination With Ursodeoxycholic Acid in Primary Biliary Cirrhosis (BEZURSO) trial, are currently awaited. SUMMARY A considerable body of observational evidence supports the safety and efficacy of fibrate treatment in PBC patients with an incomplete response to UDCA. These results encourage the evaluation of its effects on liver-related morbidity and mortality in larger clinical trials.
Collapse
|
139
|
Tsai TH, Shih SC, Ho TC, Ma HI, Liu MY, Chen SL, Tsao YP. Pigment epithelium-derived factor 34-mer peptide prevents liver fibrosis and hepatic stellate cell activation through down-regulation of the PDGF receptor. PLoS One 2014; 9:e95443. [PMID: 24763086 PMCID: PMC3998957 DOI: 10.1371/journal.pone.0095443] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 03/27/2014] [Indexed: 01/28/2023] Open
Abstract
Pigment epithelium-derived factor (PEDF) has been shown previously to prevent liver fibrosis and hepatic stellate cell (HSC) activation. By investigating the functional domains in PEDF, we identified a 34-mer peptide (residues Asp44-Asn77) that harbors the same function as the full-length PEDF protein. Not only did the 34-mer suppress the development of fibrosis in carbon tetrachloride (CCl4)-treated mouse liver but it also upregulated peroxisome proliferator-activated receptor-gamma (PPARγ) expression in HSCs in vivo. Platelet-derived growth factor (PDGF) plays a crucial role on the process of HSC activation in response to liver damage. The 34-mer suppressed PDGF-induced cell proliferation and expression of myofibroblastic marker proteins in primary rat HSC culture, increased the levels of PPARγ mRNA and protein in a dose-dependent manner and markedly reduced the level of active β-catenin protein, an HSC activating factor, in HSC-T6 cells. Similarly, IWR-1, an inhibitor of the Wnt response, displayed the same effect as the 34-mer in preventing HSC-T6 activation. The Wnt signaling-mediated PPARγ suppression was abolished by both the IWR-1 inhibitor and a small interfering RNA (siRNA) targeting β-catenin and the Wnt coreceptor, LRP6. Both PEDF and the 34-mer down-regulated PDGF receptor-α/β expression and blocked the PDGF-induced phosphorylation of Akt and ERK. Moreover, the inhibitory effect on PDGF receptor expression was abolished by PPARγ antagonists and PPARγ siRNA. Our observations indicate that the PEDF-derived 34-mer peptide can mimic PEDF in attenuating HSC activation. Investigation of this 34-mer peptide led to the identification of a signaling mechanism involving PPARγ induction, suppression of Wnt/β-catenin signaling and down-regulation of the PDGF receptor-α/β.
Collapse
Affiliation(s)
- Tung-Han Tsai
- Department of Neurosurgery, Tri-Service General Hospital, National Defense Center, Taipei, Republic of China
| | - Shou-Chuan Shih
- Department of Gastroenterology, Mackay Memorial Hospital, Taipai, Republic of China
- Mackay Medicine, Nursing and Management College, Taipei, Republic of China
| | - Tsung-Chuan Ho
- Department of Medical Research, Mackay Memorial Hospital, Taipei, Republic of China
| | - Hsin-I Ma
- Department of Neurosurgery, Tri-Service General Hospital, National Defense Center, Taipei, Republic of China
| | - Ming-Ying Liu
- Department of Neurosurgery, Tri-Service General Hospital, National Defense Center, Taipei, Republic of China
| | - Show-Li Chen
- Department of Microbiology, School of Medicine, National Taiwan University, Taipei, Republic of China
| | - Yeou-Ping Tsao
- Department of Medical Research, Mackay Memorial Hospital, Taipei, Republic of China
- * E-mail:
| |
Collapse
|
140
|
Firrincieli D, Braescu T, Housset C, Chignard N. Illuminating liver fibrosis with vitamin D. Clin Res Hepatol Gastroenterol 2014; 38:5-8. [PMID: 24238723 DOI: 10.1016/j.clinre.2013.10.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Revised: 09/27/2013] [Accepted: 10/10/2013] [Indexed: 02/04/2023]
Abstract
Hepatic fibrosis results from the accumulation of extracellular matrix-producing myofibroblasts in the liver. The mechanisms leading to the activation of hepatic stellate cells (HSCs) into myofibroblasts have been well described. By contrast, few molecular pathways leading to myofibroblast deactivation have been documented. Recently, the vitamin D-VDR axis has been shown to modulate HSC activity through a complex mechanism involving epigenetic modifications induced by the SMAD pathway.
Collapse
Affiliation(s)
- D Firrincieli
- Inserm UMR_S 938, CdR Saint-Antoine, 75012 Paris, France; UPMC Univ Paris 06, 75012 Paris, France
| | - T Braescu
- Inserm UMR_S 938, CdR Saint-Antoine, 75012 Paris, France; UPMC Univ Paris 06, 75012 Paris, France
| | - C Housset
- Inserm UMR_S 938, CdR Saint-Antoine, 75012 Paris, France; UPMC Univ Paris 06, 75012 Paris, France; AP-HP, hôpital Saint-Antoine, service d'hépatologie, 75012 Paris, France
| | - N Chignard
- Inserm UMR_S 938, CdR Saint-Antoine, 75012 Paris, France; UPMC Univ Paris 06, 75012 Paris, France.
| |
Collapse
|
141
|
Mansour M. The Roles of Peroxisome Proliferator-Activated Receptors in the Metabolic Syndrome. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2014; 121:217-66. [DOI: 10.1016/b978-0-12-800101-1.00007-7] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
142
|
Staels B, Rubenstrunk A, Noel B, Rigou G, Delataille P, Millatt LJ, Baron M, Lucas A, Tailleux A, Hum DW, Ratziu V, Cariou B, Hanf R. Hepatoprotective effects of the dual peroxisome proliferator-activated receptor alpha/delta agonist, GFT505, in rodent models of nonalcoholic fatty liver disease/nonalcoholic steatohepatitis. Hepatology 2013; 58:1941-52. [PMID: 23703580 DOI: 10.1002/hep.26461] [Citation(s) in RCA: 330] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Revised: 03/11/2013] [Accepted: 04/10/2013] [Indexed: 12/11/2022]
Abstract
UNLABELLED Nonalcoholic fatty liver disease (NAFLD) covers a spectrum of liver damage ranging from simple steatosis to nonalcoholic steatohepatitis (NASH), fibrosis, and cirrhosis. To date, no pharmacological treatment is approved for NAFLD/NASH. Here, we report on preclinical and clinical data with GFT505, a novel dual peroxisome proliferator-activated receptor alpha/delta (PPAR-α/δ) agonist. In the rat, GFT505 concentrated in the liver with limited extrahepatic exposure and underwent extensive enterohepatic cycling. The efficacy of GFT505 was assessed in animal models of NAFLD/NASH and liver fibrosis (Western diet [WD]-fed human apolipoprotein E2 [hApoE2] transgenic mice, methionine- and choline-deficient diet-fed db/db mice, and CCl4 -induced fibrosis in rats). GFT505 demonstrated liver-protective effects on steatosis, inflammation, and fibrosis. In addition, GFT505 improved liver dysfunction markers, decreased hepatic lipid accumulation, and inhibited proinflammatory (interleukin-1 beta, tumor necrosis factor alpha, and F4/80) and profibrotic (transforming growth factor beta, tissue inhibitor of metalloproteinase 2, collagen type I, alpha 1, and collagen type I, alpha 2) gene expression. To determine the role of PPAR-α-independent mechanisms, the effect of GFT505 was assessed in hApoE2 knock-in/PPAR-α knockout mice. In these mice, GFT505 also prevented WD-induced liver steatosis and inflammation, indicating a contribution of PPAR-α-independent mechanisms. Finally, the effect of GFT505 on liver dysfunction markers was assessed in a combined analysis of four phase II clinical studies in metabolic syndrome patients. GFT505 treatment decreased plasma concentrations of alanine aminotransferase, gamma-glutamyl transpeptidase, and alkaline phosphatase. CONCLUSION The dual PPAR-α/δ agonist, GFT505, is a promising liver-targeted drug for treatment of NAFLD/NASH. In animals, its protective effects are mediated by both PPAR-α-dependent and -independent mechanisms.
Collapse
Affiliation(s)
- Bart Staels
- Institut Pasteur de Lille, Lille, France; Inserm, UMR1011, Lille, France; Université Lille Nord de France, Lille, France; Université Droit et Santé de Lille, Lille, France
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
143
|
Kumar V, Mundra V, Mahato RI. Nanomedicines of Hedgehog inhibitor and PPAR-γ agonist for treating liver fibrosis. Pharm Res 2013; 31:1158-69. [PMID: 24249038 DOI: 10.1007/s11095-013-1239-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Accepted: 10/20/2013] [Indexed: 01/01/2023]
Abstract
PURPOSE Hedgehog (Hh) and peroxisome proliferator-activated receptor gamma (PPAR-γ) are major signaling pathways involved in the pathogenesis of liver fibrosis. Since Hh inhibitor, vismodegib (GDC) and PPAR-γ agonist, rosiglitazone (RSG) have poor water solubility, our objective was to formulate biodegradable polymeric nanoparticles encapsulating GDC and RSG for treating liver fibrosis. METHODS Methoxy-polyethylene-glycol-b-poly(carbonate-co-lactide) [mPEG-b-p(CB-co-LA)] was synthesized and characterized using (1)H NMR. Nanoparticles were prepared using this polymer by emulsification/solvent evaporation method to encapsulate GDC and RSG either alone or in combination. Nanoparticles were characterized for particle size, drug loading, drug release, and anti-fibrotic efficacy after tail vein injection into common bile duct ligated (CBDL) fibrotic rats. RESULTS mPEG-b-p(CB-co-LA) copolymer has molecular weight of 30,000 Da as determined by (1)H NMR. Nanoparticles were monodisperse with a mean particle size of 120-130 nm. Drug loading was 5% and 2% w/w for GDC and RSG, respectively. Nanoparticles carrying both GDC and RSG were formulated at half of their individual drug loading. Systemic administration of drug loaded nanoparticles protected liver injury in CBDL rats by suppressing the activation of hepatic stellate cells, and decreasing inflammatory cytokines. CONCLUSION Polymeric nanoparticles for co-delivery of Hh inhibitor and PPAR-γ agonist have the potential to treat liver fibrosis by intervening complex fibrotic cascade.
Collapse
Affiliation(s)
- Virender Kumar
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee, 38163, USA
| | | | | |
Collapse
|
144
|
Friedman SL. Focus. J Hepatol 2013; 59:915-7. [PMID: 23958934 DOI: 10.1016/j.jhep.2013.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Accepted: 08/12/2013] [Indexed: 12/04/2022]
Affiliation(s)
- Scott L Friedman
- Division of Liver Diseases, Mount Sinai School of Medicine, New York, NY, United States.
| |
Collapse
|
145
|
Morán-Salvador E, Titos E, Rius B, González-Périz A, García-Alonso V, López-Vicario C, Miquel R, Barak Y, Arroyo V, Clària J. Cell-specific PPARγ deficiency establishes anti-inflammatory and anti-fibrogenic properties for this nuclear receptor in non-parenchymal liver cells. J Hepatol 2013; 59:1045-53. [PMID: 23831119 DOI: 10.1016/j.jhep.2013.06.023] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Revised: 06/19/2013] [Accepted: 06/24/2013] [Indexed: 01/11/2023]
Abstract
BACKGROUND & AIMS PPARγ plays an essential role in the transcriptional regulation of genes involved in lipid and glucose metabolism, insulin sensitivity, and inflammation. We recently demonstrated that PPARγ plays a causative role in hepatocyte lipid deposition, contributing to the pathogenesis of hepatic steatosis. In this study, we investigated the role of PPARγ in the inflammatory and fibrogenic response of the liver. METHODS Heterozygous floxed/null Cre/LoxP mice with targeted deletion of PPARγ in either hepatocytes (Alb-Cre), macrophages (LysM-Cre) or hepatic stellate cells (HSCs) (aP2-Cre) were submitted to carbon tetrachloride (CCl4) liver injury. Further analyses were performed in precision-cut liver slices (PCLS) and primary cultures of hepatocytes, macrophages, and HSCs. RESULTS LysM-Cre mice displayed an exacerbated response to chronic CCl4 injury and showed higher necroinflammatory injury, lipid peroxidation, inflammatory infiltrate, cleaved-caspase-3 and caspase 3/7 activity, and COX-2, TNF-α, CXCL2, and IL-1β expression than Alb-Cre and control mice. The deleterious effects of PPARγ disruption in liver macrophages were confirmed in an acute model of CCl4 injury as well as in PCLS incubated with LPS. Moreover, LysM-Cre mice showed an aggravated fibrogenic response to CCl4, as revealed by more prominent Sirius Red and Masson's trichrome staining, elevated hydroxyproline content and induced α-SMA and TIMP-1 expression. Importantly, aP2-Cre mice with specific disruption of PPARγ in HSCs, as confirmed by immunocytochemical analysis of individual liver cells, also showed exacerbated liver damage and fibrogenic response to CCl4. CONCLUSIONS These data unveil anti-inflammatory and anti-fibrogenic roles for PPARγ in non-parenchymal liver cells.
Collapse
Affiliation(s)
- Eva Morán-Salvador
- Department of Biochemistry and Molecular Genetics, Hospital Clínic-IDIBAPS-Esther Koplowitz Center, Barcelona, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
146
|
Oldenburg AR, Delbarre E, Thiede B, Vigouroux C, Collas P. Deregulation of Fragile X-related protein 1 by the lipodystrophic lamin A p.R482W mutation elicits a myogenic gene expression program in preadipocytes. Hum Mol Genet 2013; 23:1151-62. [PMID: 24108105 DOI: 10.1093/hmg/ddt509] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The nuclear lamina is implicated in the regulation of various nuclear functions. Several laminopathy-causing mutations in the LMNA gene, notably the p.R482W substitution linked to familial partial lipodystrophy type 2 (FPLD2), are clustered in the immunoglobulin fold of lamin A. We report a functional association between lamin A and fragile X-related protein 1 (FXR1P), a protein of the fragile X-related family involved in fragile X syndrome. Searching for proteins differentially interacting with the immunoglobulin fold of wild-type and R482W mutant lamin A, we identify FXR1P as a novel component of the lamin A protein network. The p.R482W mutation abrogates interaction of FXR1P with lamin A. Fibroblasts from FPLD2 patients display elevated levels of FXR1P and delocalized FXR1P. In human adipocyte progenitors, deregulation of lamin A expression leads to FXR1P up-regulation, impairment of adipogenic differentiation and induction of myogenin expression. FXR1P overexpression also stimulates a myogenic gene expression program in these cells. Our results demonstrate a cross-talk between proteins hitherto implicated in two distinct mesodermal pathologies. We propose a model where the FPLD2 lamin A p.R482W mutation elicits, through up-regulation of FXR1P, a remodeling of an adipogenic differentiation program into a myogenic program.
Collapse
Affiliation(s)
- Anja R Oldenburg
- Stem Cell Epigenetics Laboratory, Institute of Basic Medical Sciences and Norwegian Center for Stem Cell Research, Faculty of Medicine, University of Oslo, PO Box 1112, Blindern, Oslo 0317, Norway
| | | | | | | | | |
Collapse
|
147
|
Sharvit E, Abramovitch S, Reif S, Bruck R. Amplified inhibition of stellate cell activation pathways by PPAR-γ, RAR and RXR agonists. PLoS One 2013; 8:e76541. [PMID: 24098526 PMCID: PMC3788137 DOI: 10.1371/journal.pone.0076541] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2013] [Accepted: 08/28/2013] [Indexed: 02/06/2023] Open
Abstract
Peroxisome proliferator activator receptors (PPAR) ligands such as 15-Δ12,13-prostaglandin L(2) [PJ] and all trans retinoic acid (ATRA) have been shown to inhibit the development of liver fibrosis. The role of ligands of retinoic X receptor (RXR) and its ligand, 9-cis, is less clear. The purpose of this study was to investigate the effects of combined treatment of the three ligends, PJ, ATRA and 9-cis, on key events during liver fibrosis in rat primary hepatic stellate cells (HSCs). We found that the anti-proliferative effect of the combined treatment of PJ, ATRA and 9-cis on HSCs was additive. Further experiments revealed that this inhibition was due to cell cycle arrest at the G0/G1 phase as demonstrated by FACS analysis. In addition, the combined treatment reduced cyclin D1 expression and increased p21 and p27 protein levels. Furthermore, we found that the three ligands down regulated the phosphorylation of mTOR and p70(S6K). The activation of HSCs was also inhibited by the three ligands as shown by inhibition of vitamin A lipid droplets depletion from HSCs. Studies using real time PCR and western blot analysis showed marked inhibition of collagen Iα1 and αSMA by the combination of the three ligands. These findings suggest that the combined use of PJ, ATRA and 9-cis causes inhibition of cell proliferation by cell cycle arrest and down-regulation of fibrotic markers to a greater extent compared to each of the ligands alone.
Collapse
Affiliation(s)
- Efrat Sharvit
- Institute of Gastroenterology and Liver Diseases, Tel Aviv Sourasky Medical Center, and Sackler Faculty of Medicine, Tel Aviv, Israel
- Pediatric Gastroenterology Unit, Dana Children’s Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shirley Abramovitch
- Pediatric Gastroenterology Unit, Dana Children’s Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Shimon Reif
- Institute of Gastroenterology and Liver Diseases, Tel Aviv Sourasky Medical Center, and Sackler Faculty of Medicine, Tel Aviv, Israel
- Pediatric Gastroenterology Unit, Dana Children’s Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Rafael Bruck
- Institute of Gastroenterology and Liver Diseases, Tel Aviv Sourasky Medical Center, and Sackler Faculty of Medicine, Tel Aviv, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
148
|
Chen A, Tang Y, Davis V, Hsu FF, Kennedy SM, Song H, Turk J, Brunt EM, Newberry EP, Davidson NO. Liver fatty acid binding protein (L-Fabp) modulates murine stellate cell activation and diet-induced nonalcoholic fatty liver disease. Hepatology 2013; 57:2202-12. [PMID: 23401290 PMCID: PMC3665693 DOI: 10.1002/hep.26318] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2012] [Accepted: 01/31/2013] [Indexed: 12/12/2022]
Abstract
UNLABELLED Activation of hepatic stellate cells (HSCs) is crucial to the development of fibrosis in nonalcoholic fatty liver disease. Quiescent HSCs contain lipid droplets (LDs), whose depletion upon activation induces a fibrogenic gene program. Here we show that liver fatty acid-binding protein (L-Fabp), an abundant cytosolic protein that modulates fatty acid (FA) metabolism in enterocytes and hepatocytes, also modulates HSC FA utilization and in turn regulates the fibrogenic program. L-Fabp expression decreased 10-fold following HSC activation, concomitant with depletion of LDs. Primary HSCs isolated from L-FABP(-/-) mice contain fewer LDs than wild-type (WT) HSCs, and exhibit up-regulated expression of genes involved in HSC activation. Adenoviral L-Fabp transduction inhibited activation of passaged WT HSCs and increased both the expression of prolipogenic genes and also augmented intracellular lipid accumulation, including triglyceride and FA, predominantly palmitate. Freshly isolated HSCs from L-FABP(-/-) mice correspondingly exhibited decreased palmitate in the free FA pool. To investigate whether L-FABP deletion promotes HSC activation in vivo, we fed L-FABP(-/-) and WT mice a high-fat diet supplemented with trans-fatty acids and fructose (TFF). TFF-fed L-FABP(-/-) mice exhibited reduced hepatic steatosis along with decreased LD abundance and size compared to WT mice. In addition, TFF-fed L-FABP(-/-) mice exhibited decreased hepatic fibrosis, with reduced expression of fibrogenic genes, compared to WT mice. CONCLUSION L-FABP deletion attenuates both diet-induced hepatic steatosis and fibrogenesis, despite the observation that L-Fabp paradoxically promotes FA and LD accumulation and inhibits HSC activation in vitro. These findings highlight the importance of cell-specific modulation of hepatic lipid metabolism in promoting fibrogenesis in nonalcoholic fatty liver disease. (Hepatology 2013).
Collapse
Affiliation(s)
- Anping Chen
- Departments of Pathology, School of Medicine, Saint Louis University, Saint Louis, MO. 63104
| | - Youcai Tang
- Departments of Pathology, School of Medicine, Saint Louis University, Saint Louis, MO. 63104
| | - Victoria Davis
- Department of Medicine, Washington University School of Medicine, St. Louis, MO. 63110
| | - Fong-Fu Hsu
- Department of Medicine, Washington University School of Medicine, St. Louis, MO. 63110
| | - Susan M. Kennedy
- Department of Medicine, Washington University School of Medicine, St. Louis, MO. 63110
| | - Haowei Song
- Department of Medicine, Washington University School of Medicine, St. Louis, MO. 63110
| | - John Turk
- Department of Medicine, Washington University School of Medicine, St. Louis, MO. 63110
| | - Elizabeth M. Brunt
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO. 63110
| | - Elizabeth P. Newberry
- Department of Medicine, Washington University School of Medicine, St. Louis, MO. 63110
| | - Nicholas O. Davidson
- Department of Medicine, Washington University School of Medicine, St. Louis, MO. 63110,Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO. 63110,Corresponding author: Tel: 314-362-2027; Fax: 314-362-2033;
| |
Collapse
|
149
|
Bian EB, Zhao B, Huang C, Wang H, Meng XM, Wu BM, Ma TT, Zhang L, Lv XW, Li J. New advances of DNA methylation in liver fibrosis, with special emphasis on the crosstalk between microRNAs and DNA methylation machinery. Cell Signal 2013; 25:1837-44. [PMID: 23707524 DOI: 10.1016/j.cellsig.2013.05.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 05/07/2013] [Indexed: 12/17/2022]
Abstract
Epigenetics refers to the study of heritable changes in the pattern of gene expression that is controlled by a mechanism specifically not due to changes the primary DNA sequence. Well-known epigenetic mechanisms include DNA methylation, post-translational histone modifications and RNA-based mechanisms including those controlled by small non-coding RNAs (miRNAs). Recent studies have shown that epigenetic modifications orchestrate the hepatic stellate cell (HSC) activation and liver fibrosis. In this review we focus on the aberrant methylation of CpG island promoters of select genes is the prominent epigenetic mechanism to effectively silence gene transcription facilitating HSC activation and liver fibrosis. Furthermore, we also discuss epigenetic dysregulation of tumor-suppressor miRNA genes by promoter DNA methylation and the interaction of DNA methylation with miRNAs involved in the regulation of HSC activation and liver fibrosis. Recent advances in epigenetics alterations in the pathogenesis of liver fibrosis and their possible use as new therapeutic targets and biomarkers.
Collapse
Affiliation(s)
- Er-Bao Bian
- Institute for Liver Diseases of Anhui Medical University, Hefei 230032, Anhui Province, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
150
|
Mezaki Y, Morii M, Hebiguchi T, Yoshikawa K, Yamaguchi N, Yoshino H, Senoo H. The role of retinoic acid receptors in activated hepatic stellate cells. Med Hypotheses 2013; 81:222-4. [PMID: 23688744 DOI: 10.1016/j.mehy.2013.04.045] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Accepted: 04/26/2013] [Indexed: 02/02/2023]
Abstract
Hepatic stellate cells (HSCs), also known as Ito cells, fat-storing cells, vitamin A-storing cells or lipocytes, reside in the spaces between hepatocytes and liver sinusoids. Vitamin A storage within the HSCs is achieved through the cooperative action of two proteins, cellular retinol-binding protein (CRBP) I and lecithin:retinol acyltransferase (LRAT). After the discovery that HSCs are responsible not only for the storage of vitamin A, but also for the development of liver fibrosis and subsequent liver cirrhosis, HSCs have been considered a therapeutic target for prevention or reversal of liver fibrogenesis. We have reported that HSCs acquire retinoid responsiveness after in vitro activation by post-transcriptional upregulation of retinoic acid receptor α gene expression. Here we extend this observation in relation to the functions of CRBP I and LRAT, and propose a hypothesis that increased retinoid signaling in activated HSCs forms a feedback loop toward vitamin A restoration in the liver.
Collapse
Affiliation(s)
- Yoshihiro Mezaki
- Department of Cell Biology and Morphology, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita 010-8543, Japan.
| | | | | | | | | | | | | |
Collapse
|