51
|
Marí M, Morales A, Colell A, García-Ruiz C, Fernández-Checa JC. Mitochondrial cholesterol accumulation in alcoholic liver disease: Role of ASMase and endoplasmic reticulum stress. Redox Biol 2014; 3:100-8. [PMID: 25453982 PMCID: PMC4297930 DOI: 10.1016/j.redox.2014.09.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 09/21/2014] [Accepted: 09/23/2014] [Indexed: 02/08/2023] Open
Abstract
Alcoholic liver disease (ALD) is a major cause of chronic liver disease and a growing health concern in theworld. While the pathogenesis of ALD is poorly characterized key players identified in experimental models and patients, such as perturbations in mitochondrial structure and function, selective loss of antioxidant defense and susceptibility to inflammatory cytokines, contribute to ALD progression. Both oxidative stress and mitochondrial dysfunction compromise essential cellular functions and energy generation and hence are important pathogenic mechanisms of ALD. An important process mediating the mitochondrial disruption induced by alcohol intake is the trafficking of cholesterol to mitochondria, mediated by acid sphingomyelinase-induced endoplasmic reticulum stress, which contributes to increased cholesterol synthesis and StARD1upregulation. Mitochondrial cholesterol accumulation not only sensitizes to oxidative stress but it can contribute to the metabolic reprogramming in ALD, manifested by activation of the hypoxia inducible transcription factor 1 and stimulation of glycolysis and lactate secretion. Thus, a better understanding of the mechanisms underlying alcohol-mediated mitochondrial impairment and oxidative stress may lead to the identification of novel treatments for ALD. The present review briefly summarizes current knowledge on the cellular and molecular mechanisms contributing to alcohol-induced mitochondrial dysfunction and cholesterol accumulation and provides insights for potential therapeutic targets in ALD. Alcohol perturbs mitochondria function, which modulates ROS generation and alcohol metabolism. Alcohol stimulates mitochondrial cholesterol (mChol) accumulation. MChol accumulation impairs mitochondrial function and mediates alcohol-induced lipotoxicity. ASMase promotes mitochondrial dysfunction by stimulating mChol loading.
Collapse
Affiliation(s)
- Montserrat Marí
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB-CSIC), Consejo Superior Investigaciones Científicas (CSIC), IDIBAPS, Liver Unit-Hospital Clínic, CIBEREHD, 08036 Barcelona, Spain.
| | - Albert Morales
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB-CSIC), Consejo Superior Investigaciones Científicas (CSIC), IDIBAPS, Liver Unit-Hospital Clínic, CIBEREHD, 08036 Barcelona, Spain
| | - Anna Colell
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB-CSIC), Consejo Superior Investigaciones Científicas (CSIC), IDIBAPS, Liver Unit-Hospital Clínic, CIBEREHD, 08036 Barcelona, Spain
| | - Carmen García-Ruiz
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB-CSIC), Consejo Superior Investigaciones Científicas (CSIC), IDIBAPS, Liver Unit-Hospital Clínic, CIBEREHD, 08036 Barcelona, Spain
| | - Jose C Fernández-Checa
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB-CSIC), Consejo Superior Investigaciones Científicas (CSIC), IDIBAPS, Liver Unit-Hospital Clínic, CIBEREHD, 08036 Barcelona, Spain; Research Center for Alcoholic Liver and Pancreatic Diseases, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
52
|
Hendrikx T, Walenbergh SMA, Hofker MH, Shiri-Sverdlov R. Lysosomal cholesterol accumulation: driver on the road to inflammation during atherosclerosis and non-alcoholic steatohepatitis. Obes Rev 2014; 15:424-33. [PMID: 24629059 DOI: 10.1111/obr.12159] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Revised: 01/05/2014] [Accepted: 01/05/2014] [Indexed: 12/15/2022]
Abstract
Many studies show an association between the accumulation of cholesterol inside lysosomes and the progression towards inflammatory disease states that are closely related to obesity. While in the past, the knowledge regarding lysosomal cholesterol accumulation was limited to its association with plaque severity during atherosclerosis, recently, a growing body of evidence indicates a causal link between lysosomal cholesterol accumulation and inflammation. These findings make lysosomal cholesterol accumulation an important target for intervention in metabolic diseases that are characterized by the presence of an inflammatory response. In this review, we aim to show the importance of cholesterol trapping inside lysosomes to the development of inflammation by focusing upon cardiovascular disease and non-alcoholic steatohepatitis (NASH) in particular. We summarize current data supporting the hypothesis that lysosomal cholesterol accumulation plays a key role in the development of inflammation during atherosclerosis and NASH. In addition, potential mechanisms by which disturbed lysosomal function can trigger the inflammatory response, the challenges in improving cholesterol trafficking in macrophages and recent successful research directions will be discussed.
Collapse
Affiliation(s)
- T Hendrikx
- Department of Molecular Genetics, Maastricht University, Maastricht, The Netherlands
| | | | | | | |
Collapse
|
53
|
Kasabova M, Joulin-Giet A, Lecaille F, Gilmore BF, Marchand-Adam S, Saidi A, Lalmanach G. Regulation of TGF-β1-driven differentiation of human lung fibroblasts: emerging roles of cathepsin B and cystatin C. J Biol Chem 2014; 289:16239-51. [PMID: 24790080 DOI: 10.1074/jbc.m113.542407] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Lung matrix homeostasis partly depends on the fine regulation of proteolytic activities. We examined the expression of human cysteine cathepsins (Cats) and their relative contribution to TGF-β1-induced fibroblast differentiation into myofibroblasts. Assays were conducted using both primary fibroblasts obtained from patients with idiopathic pulmonary fibrosis and human lung CCD-19Lu fibroblasts. Pharmacological inhibition and genetic silencing of Cat B diminished α-smooth muscle actin expression, delayed fibroblast differentiation, and led to an accumulation of intracellular 50-kDa TGF-β1. Moreover, the addition of Cat B generated a 25-kDa mature form of TGF-β1 in Cat B siRNA-pretreated lysates. Inhibition of Cat B decreased Smad 2/3 phosphorylation but had no effect on p38 MAPK and JNK phosphorylation, indicating that Cat B mostly disturbs TGF-β1-driven canonical Smad signaling pathway. Although mRNA expression of cystatin C was stable, its secretion, which was inhibited by brefeldin A, increased during TGF-β1-induced differentiation of idiopathic pulmonary fibrosis and CCD-19Lu fibroblasts. In addition, cystatin C participated in the control of extracellular Cats, because its gene silencing restored their proteolytic activities. These data support the notion that Cat B participates in lung myofibrogenesis as suggested for stellate cells during liver fibrosis. Moreover, we propose that TGF-β1 promotes fibrosis by driving the effective cystatin C-dependent inhibition of extracellular matrix-degrading Cats.
Collapse
Affiliation(s)
- Mariana Kasabova
- From the INSERM U1100, Pathologies Pulmonaires: Protéolyse et Aérosolthérapie, Equipe 2: Mécanismes Protéolytiques dans l'Inflammation, Centre d'Etude des Pathologies Respiratoires, Université François Rabelais, Faculté de Médecine, F-37032 Tours, France and
| | - Alix Joulin-Giet
- From the INSERM U1100, Pathologies Pulmonaires: Protéolyse et Aérosolthérapie, Equipe 2: Mécanismes Protéolytiques dans l'Inflammation, Centre d'Etude des Pathologies Respiratoires, Université François Rabelais, Faculté de Médecine, F-37032 Tours, France and
| | - Fabien Lecaille
- From the INSERM U1100, Pathologies Pulmonaires: Protéolyse et Aérosolthérapie, Equipe 2: Mécanismes Protéolytiques dans l'Inflammation, Centre d'Etude des Pathologies Respiratoires, Université François Rabelais, Faculté de Médecine, F-37032 Tours, France and
| | - Brendan F Gilmore
- the Queen's University Belfast, School of Pharmacy, McClay Research Centre, Belfast, BT9 7BL, United Kingdom
| | - Sylvain Marchand-Adam
- From the INSERM U1100, Pathologies Pulmonaires: Protéolyse et Aérosolthérapie, Equipe 2: Mécanismes Protéolytiques dans l'Inflammation, Centre d'Etude des Pathologies Respiratoires, Université François Rabelais, Faculté de Médecine, F-37032 Tours, France and
| | - Ahlame Saidi
- From the INSERM U1100, Pathologies Pulmonaires: Protéolyse et Aérosolthérapie, Equipe 2: Mécanismes Protéolytiques dans l'Inflammation, Centre d'Etude des Pathologies Respiratoires, Université François Rabelais, Faculté de Médecine, F-37032 Tours, France and
| | - Gilles Lalmanach
- From the INSERM U1100, Pathologies Pulmonaires: Protéolyse et Aérosolthérapie, Equipe 2: Mécanismes Protéolytiques dans l'Inflammation, Centre d'Etude des Pathologies Respiratoires, Université François Rabelais, Faculté de Médecine, F-37032 Tours, France and
| |
Collapse
|
54
|
Kim JB, Ann YH, Park SY, Jee HG, Kim HR, Lee JH, Yu SJ, Lee HS, Kim YJ. Side population in LX2 cells decreased by transforming growth factor-β. Hepatol Res 2014; 44:229-37. [PMID: 23578133 DOI: 10.1111/hepr.12106] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Revised: 02/18/2013] [Accepted: 02/25/2013] [Indexed: 12/11/2022]
Abstract
AIM Side population (SP) cells are known to be enriched in stem/progenitor-like cells. Transforming growth factor (TGF)-β signaling is associated with extracellular matrix (ECM) production in hepatic stellate cells. We hypothesized that the SP fraction in LX2 cells is associated with ECM deposition, which is regulated through TGF-β signaling. METHODS We investigated the relationship between SP cells and TGF-β signaling in the hepatic stellate cell line LX2. The effects of TGF-β and SB431542 on the SP fraction and expression of collagen type I and phospho-Smad2 was determined. RESULTS We identified 0.8-3% SP cells in LX2 cells. The growth rate of sorted SP and non-SP cells was similar to that of the original LX2 population, but population of the G0/G1 phase was increased in SP cells. Treatment of LX2 cells with TGF-β decreased the SP fraction in a dose-dependent manner and increased the production of collagen type I. Treatment of LX2 cells with SB431542 blocked the effect of TGF-β on the SP fraction and the expression of collagen type I. We cultured LX2 cells on collagen-coated dishes to observe the effect of ECM deposition on the SP fraction. The growth rate and cell cycle distribution was similar to that observed on normal tissue culture dishes, but the SP fraction was decreased when LX2 cells were cultured on collagen-coated plates. CONCLUSION These results show that LX2 cells contain an SP fraction and that TGF-β signaling is involved in the induction of ECM deposition as well as the number of SP cells.
Collapse
Affiliation(s)
- Jong Bin Kim
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
55
|
Kong XY, Nesset CK, Damme M, Løberg EM, Lübke T, Mæhlen J, Andersson KB, Lorenzo PI, Roos N, Thoresen GH, Rustan AC, Kase ET, Eskild W. Loss of lysosomal membrane protein NCU-G1 in mice results in spontaneous liver fibrosis with accumulation of lipofuscin and iron in Kupffer cells. Dis Model Mech 2014; 7:351-62. [PMID: 24487409 PMCID: PMC3944495 DOI: 10.1242/dmm.014050] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Human kidney predominant protein, NCU-G1, is a highly conserved protein with an unknown biological function. Initially described as a nuclear protein, it was later shown to be a bona fide lysosomal integral membrane protein. To gain insight into the physiological function of NCU-G1, mice with no detectable expression of this gene were created using a gene-trap strategy, and Ncu-g1gt/gt mice were successfully characterized. Lysosomal disorders are mainly caused by lack of or malfunctioning of proteins in the endosomal-lysosomal pathway. The clinical symptoms vary, but often include liver dysfunction. Persistent liver damage activates fibrogenesis and, if unremedied, eventually leads to liver fibrosis/cirrhosis and death. We demonstrate that the disruption of Ncu-g1 results in spontaneous liver fibrosis in mice as the predominant phenotype. Evidence for an increased rate of hepatic cell death, oxidative stress and active fibrogenesis were detected in Ncu-g1gt/gt liver. In addition to collagen deposition, microscopic examination of liver sections revealed accumulation of autofluorescent lipofuscin and iron in Ncu-g1gt/gt Kupffer cells. Because only a few transgenic mouse models have been identified with chronic liver injury and spontaneous liver fibrosis development, we propose that the Ncu-g1gt/gt mouse could be a valuable new tool in the development of novel treatments for the attenuation of fibrosis due to chronic liver damage.
Collapse
Affiliation(s)
- Xiang Y Kong
- Department of Bioscience, University of Oslo, 0316 Oslo, Norway
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
56
|
Weerachayaphorn J, Luo Y, Mennone A, Soroka CJ, Harry K, Boyer JL. Deleterious effect of oltipraz on extrahepatic cholestasis in bile duct-ligated mice. J Hepatol 2014; 60:160-6. [PMID: 23978715 PMCID: PMC4054607 DOI: 10.1016/j.jhep.2013.08.015] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 07/18/2013] [Accepted: 08/08/2013] [Indexed: 12/30/2022]
Abstract
BACKGROUND & AIMS Oltipraz (4-methyl-5(pyrazinyl-2)-1-2-dithiole-3-thione), a promising cancer preventive agent, has an antioxidative activity and ability to enhance glutathione biosynthesis, phase II detoxification enzymes and multidrug resistance-associated protein-mediated efflux transporters. Oltipraz can protect against hepatotoxicity caused by carbon tetrachloride, acetaminophen and alpha-naphthylisothiocyanate. Whether oltipraz has hepato-protective effects on obstructive cholestasis is unknown. METHODS We administered oltipraz to mice for 5 days prior to bile duct ligation (BDL) for 3 days. Liver histology, liver function markers, bile flow rates and hepatic expression of profibrogenic genes were evaluated. RESULTS Mice pretreated with oltipraz prior to BDL demonstrated higher levels of serum aminotransferases and more severe liver damage than in control mice. Higher bile flow and glutathione secretion rates were observed in unoperated mice treated with oltipraz than in control mice, suggesting that liver necrosis in oltipraz-treated BDL mice may be related partially to increased bile-acid independent flow and biliary pressure. Oltipraz treatment in BDL mice enhanced α-smooth muscle actin expression, consistent with activation of hepatic stellate cells and portal fibroblasts. Matrix metalloproteinases (Mmp) 9 and 13 and tissue inhibitors of metalloproteinases (Timp) 1 and 2 levels were increased in the oltipraz-treated BDL group, suggesting that the secondary phase of liver injury induced by oltipraz might be due to excessive Mmp and Timp secretions, which induce remodeling of the extracellular matrix. CONCLUSIONS Oltipraz treatment exacerbates the severity of liver injury following BDL and should be avoided as therapy for extrahepatic cholestatic disorders due to bile duct obstruction.
Collapse
Affiliation(s)
- Jittima Weerachayaphorn
- Department of Internal Medicine, School of Medicine, Yale University, New Haven, Connecticut, USA,Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Yuhuan Luo
- Department of Internal Medicine, School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Albert Mennone
- Department of Internal Medicine, School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Carol J. Soroka
- Department of Internal Medicine, School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Kathy Harry
- Department of Internal Medicine, School of Medicine, Yale University, New Haven, Connecticut, USA
| | - James L. Boyer
- Department of Internal Medicine, School of Medicine, Yale University, New Haven, Connecticut, USA,The corresponding author: James L. Boyer, M.D., Emeritus Director, Liver Center, Yale University School of Medicine, 333 Cedar Street, 1080 LMP, New Haven, CT 06520. Phone: (203) 785-5279; Fax: (203) 785-7273;
| |
Collapse
|
57
|
Mousavi SA, Fønhus MS, Kindberg GM, Tolleshaug H, Berg T. Enhanced activity of lysosomal proteases in activated rat hepatic stellate cells is associated with a concomitant increase in the number of the mannose-6-phosphate/insulin-like growth factor II receptor. Cell Biol Int 2013; 37:703-712. [PMID: 23495048 DOI: 10.1002/cbin.10085] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2012] [Accepted: 02/13/2013] [Indexed: 11/08/2023]
Abstract
Activated hepatic stellate cells (HSCs) play a central role during hepatic tissue repair through their influence on extracellular matrix remodelling. We have determined whether the activity levels of cathepsin B and D are affected by in vitro activation of rat HSCs, and whether the enzymes were released from the cells. Furthermore, given the important role of the mannose-6-phosphate/insulin-like growth factor II receptor (M6P/IGF-IIR) in the intracellular transport of lysosomal enzymes, we have examined whether changes in the activity of these proteases were associated with parallel changes in the level of the M6P/IGF-IIR. The activity of cathepsin B and D increased ∼4 times between 2 and 8 days of HSC culture. This result was supported by analysing mRNA expression by RT-PCR. The cells released the enzymes into the culture medium, amounting to ∼10% of the cell-associated activity over 24 h. The release of enzymes was not affected by reducing medium pH from 7.4 to 6.2, indicating that the enzymes were transported to the medium independently of the M6P/IGF-II-R. The released cathepsin B was mostly in the inactive proenzyme form. HSC activation led to a particularly large increase in M6P/IGF-IIR expression. A large proportion of the receptors was located on the cell surface and was found to be very suitable for measuring endocytosis of (125) I-IGF-II. The results show that the endocytic activity increased in parallel with the increase in surface receptors and activity of lysosomal enzymes. Degradation of the ligand was reduced by inhibitors of lysosomal proteases and therefore took place in lysosomes.
Collapse
Affiliation(s)
- Seyed Ali Mousavi
- Department of Immunology and Transfusion Medicine, Akershus University Hospital, University of Oslo, Oslo, Norway
| | | | | | | | | |
Collapse
|
58
|
Chen H, Wang J, Xiang MX, Lin Y, He A, Jin CN, Guan J, Sukhova GK, Libby P, Wang JA, Shi GP. Cathepsin S-mediated fibroblast trans-differentiation contributes to left ventricular remodelling after myocardial infarction. Cardiovasc Res 2013; 100:84-94. [PMID: 23771947 DOI: 10.1093/cvr/cvt158] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Extracellular matrix (ECM) turnover plays an important role in left ventricular (LV) remodelling following myocardial infarction (MI). Cysteinyl cathepsins contribute to ECM catabolism in arterial diseases, suggesting their participation in post-MI remodelling. METHODS AND RESULTS Left anterior descending artery ligation-induced MI in mice showed increased expression and activity of cathepsin S (CatS). Administration of a non-selective cathepsin inhibitor, E64d, aggravated LV dysfunction at 7 and 28 days post-MI. Mechanistic studies showed that E64d increased post-MI inflammatory cell accumulation and cytokine expression, but did not affect apoptosis or angiogenesis in infarcted myocardium. Furthermore, E64d suppressed TGF-β1-induced Smad2 and Smad3 activation and expression of fibronectin extra domain A (ED-A), an alternatively spliced fibronectin variant, and subsequently prevented cardiac fibroblast trans-differentiation into myofibroblast, which contributed to post-MI collagen and fibronectin synthesis and deposition. Consistently, selective inhibition or genetically determined deficiency of CatS also reduced myocardial Smad2 and Smad3 activation and ED-A fibronectin expression, thus suppressing fibroblast trans-differentiation and resulting in adverse collagen turnover and impaired cardiac function-recapitulating the findings in mice treated with E64d. CONCLUSION Along with its established activities in ECM degradation, CatS plays novel roles in TGF-β1 signalling, myofibroblast trans-differentiation, and ECM protein synthesis, thereby regulating scar formation in the infarcted myocardium and preserving LV function after experimental MI.
Collapse
Affiliation(s)
- Han Chen
- Department of Cardiology, Cardiovascular Key Lab of Zhejiang Province, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
59
|
Noda S, Asano Y, Akamata K, Aozasa N, Taniguchi T, Takahashi T, Ichimura Y, Toyama T, Sumida H, Yanaba K, Tada Y, Sugaya M, Kadono T, Sato S. A possible contribution of altered cathepsin B expression to the development of skin sclerosis and vasculopathy in systemic sclerosis. PLoS One 2012; 7:e32272. [PMID: 22384200 PMCID: PMC3285678 DOI: 10.1371/journal.pone.0032272] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2012] [Accepted: 01/24/2012] [Indexed: 11/19/2022] Open
Abstract
Cathepsin B (CTSB) is a proteolytic enzyme potentially modulating angiogenic processes and extracellular matrix remodeling. While matrix metalloproteinases are shown to be implicated in tissue fibrosis and vasculopathy associated with systemic sclerosis (SSc), the role of cathepsins in this disease has not been well studied. The aim of this study is to evaluate the roles of CTSB in SSc. Serum pro-CTSB levels were determined by enzyme-linked immunosorbent assay in 55 SSc patients and 19 normal controls. Since the deficiency of transcription factor Fli1 in endothelial cells is potentially associated with the development of SSc vasculopathy, cutaneous CTSB expression was evaluated by immunostaining in Fli1+/− and wild type mice as well as in SSc and control subjects. The effects of Fli1 gene silencing and transforming growth factor-β (TGF-β) on CTSB expression were determined by real-time PCR in human dermal microvascular endothelial cells (HDMECs) and dermal fibroblasts, respectively. Serum pro-CTSB levels were significantly higher in limited cutaneous SSc (lcSSc) and late-stage diffuse cutaneous SSc (dcSSc) patients than in healthy controls. In dcSSc, patients with increased serum pro-CTSB levels showed a significantly higher frequency of digital ulcers than those with normal levels. CTSB expression in dermal blood vessels was increased in Fli1+/− mice compared with wild type mice and in SSc patients compared with healthy controls. Consistently, Fli1 gene silencing increased CTSB expression in HDMECs. In cultured dermal fibroblasts from early dcSSc, CTSB expression was decreased compared with normal fibroblasts and significantly reversed by TGF-β1 antisense oligonucleotide. In conclusion, up-regulation of endothelial CTSB due to Fli1 deficiency may contribute to the development of SSc vasculopathy, especially digital ulcers, while reduced expression of CTSB in lesional dermal fibroblasts is likely to be associated with skin sclerosis in early dcSSc.
Collapse
Affiliation(s)
| | - Yoshihide Asano
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
- * E-mail:
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
60
|
Etiopathogenesis of nonalcoholic steatohepatitis: role of obesity, insulin resistance and mechanisms of hepatotoxicity. Int J Hepatol 2012; 2012:212865. [PMID: 22792473 PMCID: PMC3389710 DOI: 10.1155/2012/212865] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2011] [Revised: 02/26/2012] [Accepted: 04/25/2012] [Indexed: 12/14/2022] Open
Abstract
Incidence of nonalcoholic fatty liver disease is increasing with an estimated prevalence of 20-30% in developed nations. This is leading to increased incidence of chronic liver disease, cirrhosis, and hepatocellular cancer. It is critical to understand the etiology and pathogenesis of any disease to create therapeutic targets and develop new treatments. In this paper we discuss the etiology and pathogenesis of nonalcoholic steatohepatitis with special focus on obesity, role of insulin resistance, and molecular mechanisms of hepatotoxicity.
Collapse
|
61
|
Moles A, Tarrats N, Fernández-Checa JC, Marí M. Cathepsin B overexpression due to acid sphingomyelinase ablation promotes liver fibrosis in Niemann-Pick disease. J Biol Chem 2011; 287:1178-88. [PMID: 22102288 DOI: 10.1074/jbc.m111.272393] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Niemann-Pick disease (NPD) is a lysosomal storage disease caused by the loss of acid sphingomyelinase (ASMase) that features neurodegeneration and liver disease. Because ASMase-knock-out mice models NPD and our previous findings revealed that ASMase activates cathepsins B/D (CtsB/D), our aim was to investigate the expression and processing of CtsB/D in hepatic stellate cells (HSCs) from ASMase-null mice and their role in liver fibrosis. Surprisingly, HSCs from ASMase-knock-out mice exhibit increased basal level and activity of CtsB as well as its in vitro processing in culture, paralleling the enhanced expression of fibrogenic markers α-smooth muscle actin (α-SMA), TGF-β, and pro-collagen-α1(I) (Col1A1). Moreover, pharmacological inhibition of CtsB blunted the expression of α-SMA and Col1A1 and proliferation of HSCs from ASMase-knock-out mice. Consistent with the enhanced activation of CtsB in HSCs from ASMase-null mice, the in vivo liver fibrosis induced by chronic treatment with CCl(4) increased in ASMase-null compared with wild-type mice, an effect that was reduced upon CtsB inhibition. In addition to liver, the enhanced proteolytic processing of CtsB was also observed in brain and lung of ASMase-knock-out mice, suggesting that the overexpression of CtsB may underlie the phenotype of NPD. Thus, these findings reveal a functional relationship between ASMase and CtsB and that the ablation of ASMase leads to the enhanced processing and activation of CtsB. Therefore, targeting CtsB may be of relevance in the treatment of liver fibrosis in patients with NPD.
Collapse
Affiliation(s)
- Anna Moles
- Liver Unit-Hospital Clínic, Centro de Investigación Biomédica en Red en el Área Temática de Entermedades Hepáticas y Digestivas, and Department of Cell Death and Proliferation, Instituto de Investigaciones Biomédicas de Barcelona, Consejo Superior de Investigaciones Científicas, 08036 Barcelona, Spain
| | | | | | | |
Collapse
|
62
|
Abd-Elgaliel WR, Cruz-Monserrate Z, Logsdon CD, Tung CH. Molecular imaging of Cathepsin E-positive tumors in mice using a novel protease-activatable fluorescent probe. MOLECULAR BIOSYSTEMS 2011; 7:3207-3213. [PMID: 21935563 DOI: 10.1039/c1mb05215b] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
UNLABELLED The purpose of this study is to demonstrate the ability of imaging Cathepsin E (Cath E) positive tumors in living animals through selective targeting of Cath E proteolytic activity using a sensitive molecular imaging agent. METHODS A peptide-based Cath E imaging probe and a control probe were synthesized for this study. Human Cath E-positive cancer cells (MPanc96-E) were implanted subcutaneously in nude mice. Tumor-bearing mice were examined in vivo with near-infrared fluorescence (NIRF) imaging at various time points after intravenous injection of the Cath E sensing imaging probe. Excised organs and tissues of interest were further imaged ex vivo. RESULTS Upon specific Cath E proteolytic activation, the NIRF signal of the imaging probe a was converted from an optically quenched initial state to a highly fluorescent active state. Imaging probe a was able to highlight the Cath E-positive tumors as early as 24 h post injection. Fluorescent signal in tumor was 3-fold higher than background. The confined specificity of imaging probe a to tumor associated Cath E was verified by using control imaging probe b. Both in vivo and ex vivo imaging results confirmed the superior selectivity and sensitivity of imaging probe a in Cath E imaging. CONCLUSIONS The small animal studies demonstrated the capability of probe a for imaging Cath E-positive tumors. The developed optical probe could be applied in early diagnostic imaging and guiding subsequent surgical procedure.
Collapse
Affiliation(s)
- Wael R Abd-Elgaliel
- Department of Radiology, The Methodist Hospital Research Institute, Weill Cornell Medical College, 6565 Fannin Street, B5-009, Houston, TX 77030, USA
| | - Zobeida Cruz-Monserrate
- Department of Cancer Biology, University of Texas, M. D. Anderson Cancer Center, Houston, TX, USA
| | - Craig D Logsdon
- Department of Cancer Biology, University of Texas, M. D. Anderson Cancer Center, Houston, TX, USA.,Department of GI Medical Oncology, University of Texas, M. D. Anderson Cancer Center, Houston, TX, USA
| | - Ching-Hsuan Tung
- Department of Radiology, The Methodist Hospital Research Institute, Weill Cornell Medical College, 6565 Fannin Street, B5-009, Houston, TX 77030, USA
| |
Collapse
|
63
|
Trebicka J, Racz I, Siegmund SV, Cara E, Granzow M, Schierwagen R, Klein S, Wojtalla A, Hennenberg M, Huss S, Fischer HP, Heller J, Zimmer A, Sauerbruch T. Role of cannabinoid receptors in alcoholic hepatic injury: steatosis and fibrogenesis are increased in CB2 receptor-deficient mice and decreased in CB1 receptor knockouts. Liver Int 2011; 31:860-70. [PMID: 21645218 DOI: 10.1111/j.1478-3231.2011.02496.x] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Alcohol is a common cause of hepatic liver injury with steatosis and fibrosis. Cannabinoid receptors (CB) modulate steatosis, inflammation and fibrogenesis. To investigate the differences between CB(1) and CB(2) in the hepatic response to chronic alcohol intake, we examined CB knockout mice (CB(1)(-/-), CB(2)(-/-)). METHODS Eight- to 10-week-old CB(1)(-/-), CB(2)(-/-) and wild-type mice received 16% ethanol for 35 weeks. Animals receiving water served as controls. We analysed triglyceride and hydroxyproline contents in liver homogenates. mRNA levels of CBs, pro-inflammatory cytokines [tumour necrosis factor (TNF)-α, monocyte chemotactic protein (MCP)-1, interleukin (IL)-1β] and profibrotic factors [α-smooth muscle actin (α-SMA), procollagen-Ia, platelet-derived growth factor β receptor (PDGFβ-R)] were analysed by reverse transcription-polymerase chain reaction (RT-PCR). Histology (hemalaun and eosin, oil-red O, CD3, CD45R, CD45, F4/80, Sirius red) characterized hepatic steatosis, inflammation and fibrosis. Activation of lipogenic pathways, activation and proliferation of hepatic stellate cell (HSC) were assessed by western blot [fatty acid synthase (FAS), sterol regulatory element binding protein 1c (SREBP-1c), α-SMA, proliferating cell nuclear antigen (PCNA), cathepsin D]. RESULTS Hepatic mRNA levels of the respective CBs were increased in wild-type animals and in CB(1)(-/-) mice after ethanol intake. Ethanol intake in CB(2)(-/-) mice induced much higher steatosis (SREBP-1c mediated) and inflammation (B-cell predominant infiltrates) compared with wild-type animals and CB(1)(-/-) mice. HSC activation and collagen production were increased in all groups after forced ethanol intake, being most pronounced in CB(2)(-/-) mice and least pronounced in CB(1)(-/-) mice. DISCUSSION The fact that CB(2) receptor knockout mice exhibited the most pronounced liver damage after ethanol challenge indicates a protective role of CB(2) receptor expression in chronic ethanol intake. By contrast, in CB(1) knockouts, the effect of ethanol was attenuated, suggesting aggravation of fibrogenesis and SREBP-1c-mediated steatosis via CB(1) receptor expression after ethanol intake.
Collapse
Affiliation(s)
- Jonel Trebicka
- Department of Internal Medicine I, University of Bonn, Bonn, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
64
|
Tarrats N, Moles A, Morales A, García-Ruiz C, Fernández-Checa JC, Marí M. Critical role of tumor necrosis factor receptor 1, but not 2, in hepatic stellate cell proliferation, extracellular matrix remodeling, and liver fibrogenesis. Hepatology 2011; 54:319-27. [PMID: 21523796 PMCID: PMC3125435 DOI: 10.1002/hep.24388] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
UNLABELLED Tumor necrosis factor (TNF) has been implicated in the progression of many chronic liver diseases leading to fibrosis; however, the role of TNF in fibrogenesis is controversial and the specific contribution of TNF receptors to hepatic stellate cell (HSC) activation remains to be established. Using HSCs from wild-type, TNF-receptor-1 (TNFR1) knockout, TNF-receptor-2 (TNFR2) knockout, or TNFR1/R2 double-knockout (TNFR-DKO) mice, we show that loss of both TNF receptors reduced procollagen-α1(I) expression, slowed down HSC proliferation, and impaired platelet-derived growth factor (PDGF)-induced promitogenic signaling in HSCs. TNFR-DKO HSCs exhibited decreased AKT phosphorylation and in vitro proliferation in response to PDGF. These effects were reproduced in TNFR1 knockout, but not TNFR2 knockout, HSCs. In addition, matrix metalloproteinase 9 (MMP-9) expression was dependent on TNF binding to TNFR1 in primary mouse HSCs. These results were validated in the human HSC cell line, LX2, using neutralizing antibodies against TNFR1 and TNFR2. Moreover, in vivo liver damage and fibrogenesis after bile-duct ligation were reduced in TNFR-DKO and TNFR1 knockout mice, compared to wild-type or TNFR2 knockout mice. CONCLUSION TNF regulates HSC biology through its binding to TNFR1, which is required for HSC proliferation and MMP-9 expression. These data indicate a regulatory role for TNF in extracellular matrix remodeling and liver fibrosis, suggesting that targeting TNFR1 may be of benefit to attenuate liver fibrogenesis.
Collapse
Affiliation(s)
- Núria Tarrats
- IDIBAPS, Liver Unit-Hospital Clínic, CIBEREHD, and Department of Cell Death and Proliferation, IIBB-CSIC, 08036-Barcelona, Spain
| | - Anna Moles
- IDIBAPS, Liver Unit-Hospital Clínic, CIBEREHD, and Department of Cell Death and Proliferation, IIBB-CSIC, 08036-Barcelona, Spain
| | - Albert Morales
- IDIBAPS, Liver Unit-Hospital Clínic, CIBEREHD, and Department of Cell Death and Proliferation, IIBB-CSIC, 08036-Barcelona, Spain
| | - Carmen García-Ruiz
- IDIBAPS, Liver Unit-Hospital Clínic, CIBEREHD, and Department of Cell Death and Proliferation, IIBB-CSIC, 08036-Barcelona, Spain
| | - José C. Fernández-Checa
- IDIBAPS, Liver Unit-Hospital Clínic, CIBEREHD, and Department of Cell Death and Proliferation, IIBB-CSIC, 08036-Barcelona, Spain,Research Center for Alcoholic Liver and Pancreatic Diseases, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Montserrat Marí
- IDIBAPS, Liver Unit-Hospital Clínic, CIBEREHD, and Department of Cell Death and Proliferation, IIBB-CSIC, 08036-Barcelona, Spain
| |
Collapse
|
65
|
Yang JD, Nakamura I, Roberts LR. The tumor microenvironment in hepatocellular carcinoma: current status and therapeutic targets. Semin Cancer Biol 2010; 21:35-43. [PMID: 20946957 DOI: 10.1016/j.semcancer.2010.10.007] [Citation(s) in RCA: 303] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2010] [Revised: 09/14/2010] [Accepted: 10/04/2010] [Indexed: 12/16/2022]
Abstract
A growing body of literature highlights the cross-talk between tumor cells and the surrounding peri-tumoral stroma as a key modulator of the processes of hepatocarcinogenesis, epithelial mesenchymal transition (EMT), tumor invasion and metastasis. The tumor microenvironment can be broadly classified into cellular and non-cellular components. The major cellular components include hepatic stellate cells, fibroblasts, immune, and endothelial cells. These cell types produce the non-cellular components of the tumor stroma, including extracellular matrix (ECM) proteins, proteolytic enzymes, growth factors and inflammatory cytokines. The non-cellular component of the tumor stroma modulates hepatocellular carcinoma (HCC) biology by effects on cancer signaling pathways in tumor cells and on tumor invasion and metastasis. Global gene expression profiling of HCC has revealed that the tumor microenvironment is an important component in the biologic and prognostic classification of HCC. There are substantial efforts underway to develop novel drugs targeting tumor-stromal interactions. In this review, we discuss the current knowledge about the role of the tumor microenvironment in pathogenesis of HCC, the role of the tumor microenvironment in the classification of HCC and efforts to develop treatments targeting the tumor microenvironment.
Collapse
Affiliation(s)
- Ju Dong Yang
- Miles and Shirley Fiterman Center for Digestive Diseases, Mayo Clinic College of Medicine, Rochester, MN 55905, United States
| | | | | |
Collapse
|
66
|
Trebicka J, Hennenberg M, Odenthal M, Shir K, Klein S, Granzow M, Vogt A, Dienes HP, Lammert F, Reichen J, Heller J, Sauerbruch T. Atorvastatin attenuates hepatic fibrosis in rats after bile duct ligation via decreased turnover of hepatic stellate cells. J Hepatol 2010; 53:702-12. [PMID: 20633948 DOI: 10.1016/j.jhep.2010.04.025] [Citation(s) in RCA: 174] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2009] [Revised: 03/23/2010] [Accepted: 04/15/2010] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS Activation of hepatic stellate cells (HSC) and transdifferentiation to myofibroblasts following liver injury is the main culprit for hepatic fibrosis. Myofibroblasts show increased proliferation, migration, contraction, and production of extracellular matrix (ECM). In vitro, HMG-CoA reductase inhibitors (statins) inhibit proliferation and induce apoptosis of myofibroblastic HSC. To investigate the antifibrotic effects of atorvastatin in vivo we used bile duct ligated rats (BDL). METHODS BDL rats were treated with atorvastatin (15 mg/kg/d) immediately after ligation (prophylactically) or in on-going fibrosis (therapeutically). Fibrosis was assessed by hydroxyproline content and Sirius-red staining. The activation of HSC was investigated by analysis of alphaSMA expression. mRNA levels of cytokines and procollagen were analyzed by RT-PCR, and MMP-2 activity by zymography. Proliferation was assessed by expression of cathepsins (B and D), proliferating cell nuclear antigen (PCNA), and Ki67-staining. Apoptosis was characterized by caspase-3 activity, cleavage of PARP-1, and TUNEL assay. Hepatic inflammation was investigated by serum parameters and liver histology. RESULTS Prophylactic and early therapy with atorvastatin significantly attenuated fibrosis and HSC activation. Later therapy lacked significant effects on fibrosis but reduced profibrotic cytokine expression and led to a more quiescent state of HSC with less proliferation and apoptosis, while hepatic inflammation did not change. CONCLUSIONS This study shows that very early atorvastatin treatment inhibits HSC activation and fibrosis in the BDL model in vivo, while late treatment reduces HSC turnover and activity. Our findings underline that long-term studies in humans are warranted.
Collapse
Affiliation(s)
- Jonel Trebicka
- Department of Internal Medicine I, University of Bonn, Germany.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
67
|
Martinez-Outschoorn UE, Trimmer C, Lin Z, Whitaker-Menezes D, Chiavarina B, Zhou J, Wang C, Pavlides S, Martinez-Cantarin MP, Capozza F, Witkiewicz AK, Flomenberg N, Howell A, Pestell RG, Caro J, Lisanti MP, Sotgia F. Autophagy in cancer associated fibroblasts promotes tumor cell survival: Role of hypoxia, HIF1 induction and NFκB activation in the tumor stromal microenvironment. Cell Cycle 2010; 9:3515-33. [PMID: 20855962 DOI: 10.4161/cc.9.17.12928] [Citation(s) in RCA: 332] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Recently, using a co-culture system, we demonstrated that MCF7 epithelial cancer cells induce oxidative stress in adjacent cancer-associated fibroblasts, resulting in the autophagic/lysosomal degradation of stromal caveolin-1 (Cav-1). However, the detailed signaling mechanism(s) underlying this process remain largely unknown. Here, we show that hypoxia is sufficient to induce the autophagic degradation of Cav-1 in stromal fibroblasts, which is blocked by the lysosomal inhibitor chloroquine. Concomitant with the hypoxia-induced degradation of Cav-1, we see the upregulation of a number of well-established autophagy/mitophagy markers, namely LC3, ATG16L, BNIP3, BNIP3L, HIF-1α and NFκB. In addition, pharmacological activation of HIF-1α drives Cav-1 degradation, while pharmacological inactivation of HIF-1 prevents the downregulation of Cav-1. Similarly, pharmacological inactivation of NFκB--another inducer of autophagy-prevents Cav-1 degradation. Moreover, treatment with an inhibitor of glutathione synthase, namely BSO, which induces oxidative stress via depletion of the reduced glutathione pool, is sufficient to induce the autophagic degradation of Cav-1. Thus, it appears that oxidative stress mediated induction of HIF1- and NFκB-activation in fibroblasts drives the autophagic degradation of Cav-1. In direct support of this hypothesis, we show that MCF7 cancer cells activate HIF-1α- and NFκB-driven luciferase reporters in adjacent cancer-associated fibroblasts, via a paracrine mechanism. Consistent with these findings, acute knock-down of Cav-1 in stromal fibroblasts, using an siRNA approach, is indeed sufficient to induce autophagy, with the upregulation of both lysosomal and mitophagy markers. How does the loss of stromal Cav-1 and the induction of stromal autophagy affect cancer cell survival? Interestingly, we show that a loss of Cav-1 in stromal fibroblasts protects adjacent cancer cells against apoptotic cell death. Thus, autophagic cancer-associated fibroblasts, in addition to providing recycled nutrients for cancer cell metabolism, also play a protective role in preventing the death of adjacent epithelial cancer cells. We demonstrate that cancer-associated fibroblasts upregulate the expression of TIGAR in adjacent epithelial cancer cells, thereby conferring resistance to apoptosis and autophagy. Finally, the mammary fat pads derived from Cav-1 (-/-) null mice show a hypoxia-like response in vivo, with the upregulation of autophagy markers, such as LC3 and BNIP3L. Taken together, our results provide direct support for the "Autophagic Tumor Stroma Model of Cancer Metabolism", and explain the exceptional prognostic value of a loss of stromal Cav-1 in cancer patients. Thus, a loss of stromal fibroblast Cav-1 is a biomarker for chronic hypoxia, oxidative stress and autophagy in the tumor microenvironment, consistent with its ability to predict early tumor recurrence, lymph node metastasis and tamoxifen-resistance in human breast cancers. Our results imply that cancer patients lacking stromal Cav-1 should benefit from HIF-inhibitors, NFκB-inhibitors, anti-oxidant therapies, as well as autophagy/lysosomal inhibitors. These complementary targeted therapies could be administered either individually or in combination, to prevent the onset of autophagy in the tumor stromal compartment, which results in a "lethal" tumor microenvironment.
Collapse
Affiliation(s)
- Ubaldo E Martinez-Outschoorn
- Department of Medical Oncology and The Jefferson Stem Cell Biology and Regenerative Medicine Center, Thomas Jefferson University, Philadelphia, PA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
68
|
Gopinath S, Malla RR, Gondi CS, Alapati K, Fassett D, Klopfenstein JD, Dinh DH, Gujrati M, Rao JS. Co-depletion of cathepsin B and uPAR induces G0/G1 arrest in glioma via FOXO3a mediated p27 upregulation. PLoS One 2010; 5:e11668. [PMID: 20661471 PMCID: PMC2908539 DOI: 10.1371/journal.pone.0011668] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2010] [Accepted: 06/24/2010] [Indexed: 12/21/2022] Open
Abstract
Background Cathepsin B and urokinase plasminogen activator receptor (uPAR) are both known to be overexpressed in gliomas. Our previous work and that of others strongly suggest a relationship between the infiltrative phenotype of glioma and the expression of cathepsin B and uPAR. Though their role in migration and adhesion are well studied the effect of these molecules on cell cycle progression has not been thoroughly examined. Methodology/Principal Findings Cathespin B and uPAR single and bicistronic siRNA plasmids were used to downregulate these molecules in SNB19 and U251 glioma cells. FACS analysis and BrdU incorporation assay demonstrated G0/G1 arrest and decreased proliferation with the treatments, respectively. Immunoblot and immunocyto analysis demonstrated increased expression of p27Kip1 and its nuclear localization with the knockdown of cathepsin B and uPAR. These effects could be mediated by αVβ3/PI3K/AKT/FOXO pathway as observed by the decreased αVβ3 expression, PI3K and AKT phosphorylation accompanied by elevated FOXO3a levels. These results were further confirmed with the increased expression of p27Kip1 and FOXO3a when treated with Ly294002 (10 µM) and increased luciferase expression with the siRNA and Ly294002 treatments when the FOXO binding promoter region of p27Kip1 was used. Our treatment also reduced the expression of cyclin D1, cyclin D2, p-Rb and cyclin E while the expression of Cdk2 was unaffected. Of note, the Cdk2-cyclin E complex formation was reduced significantly. Conclusion/Significance Our study indicates that cathepsin B and uPAR knockdown induces G0/G1 arrest by modulating the PI3K/AKT signaling pathway and further increases expression of p27Kip1 accompanied by the binding of FOXO3a to its promoter. Taken together, our findings provide molecular mechanism for the G0/G1 arrest induced by the downregulation of cathepsin B and uPAR in SNB19 and U251 glioma cells.
Collapse
Affiliation(s)
- Sreelatha Gopinath
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, Illinois, United States of America
| | - Rama Rao Malla
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, Illinois, United States of America
| | - Christopher S. Gondi
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, Illinois, United States of America
| | - Kiranmai Alapati
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, Illinois, United States of America
| | - Daniel Fassett
- Department of Neurosurgery, University of Illinois College of Medicine at Peoria, Peoria, Illinois, United States of America
| | - Jeffrey D. Klopfenstein
- Department of Neurosurgery, University of Illinois College of Medicine at Peoria, Peoria, Illinois, United States of America
| | - Dzung H. Dinh
- Department of Neurosurgery, University of Illinois College of Medicine at Peoria, Peoria, Illinois, United States of America
| | - Meena Gujrati
- Department of Pathology, University of Illinois College of Medicine at Peoria, Peoria, Illinois, United States of America
| | - Jasti S. Rao
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, Illinois, United States of America
- Department of Neurosurgery, University of Illinois College of Medicine at Peoria, Peoria, Illinois, United States of America
- * E-mail:
| |
Collapse
|
69
|
Moles A, Tarrats N, Morales A, Domínguez M, Bataller R, Caballería J, García-Ruiz C, Fernández-Checa JC, Marí M. Acidic sphingomyelinase controls hepatic stellate cell activation and in vivo liver fibrogenesis. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 177:1214-24. [PMID: 20651240 DOI: 10.2353/ajpath.2010.091257] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The mechanisms linking hepatocellular death, hepatic stellate cell (HSC) activation, and liver fibrosis are largely unknown. Here, we investigate whether acidic sphingomyelinase (ASMase), a known regulator of death receptor and stress-induced hepatocyte apoptosis, plays a role in liver fibrogenesis. We show that selective stimulation of ASMase (up to sixfold), but not neutral sphingomyelinase, occurs during the transdifferentiation/activation of primary mouse HSCs into myofibroblast-like cells, coinciding with cathepsin B (CtsB) and D (CtsD) processing. ASMase inhibition or genetic down-regulation by small interfering RNA blunted CtsB/D processing, preventing the activation and proliferation of mouse and human HSCs (LX2 cells). In accordance, HSCs from heterozygous ASMase mice exhibited decreased CtsB/D processing, as well as lower levels of alpha-smooth muscle actin expression and proliferation. Moreover, pharmacological CtsB inhibition reproduced the antagonism of ASMase in preventing the fibrogenic properties of HSCs, without affecting ASMase activity. Interestingly, liver fibrosis induced by bile duct ligation or carbon tetrachloride administration was reduced in heterozygous ASMase mice compared with that in wild-type animals, regardless of their sensitivity to liver injury in either model. To provide further evidence for the ASMase-CtsB pathway in hepatic fibrosis, liver samples from patients with nonalcoholic steatohepatitis were studied. CtsB and ASMase mRNA levels increased eight- and threefold, respectively, in patients compared with healthy controls. These findings illustrate a novel role of ASMase in HSC biology and liver fibrogenesis by regulating its downstream effectors CtsB/D.
Collapse
Affiliation(s)
- Anna Moles
- Liver Unit, Hospital Clinic, Centre d'Investigació Biomèdica Esther Koplowitz, Centro de Investigación Biomédicas en Red en el Area temática de Enfermedades Hepáticas y Digestivas, Barcelona, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
70
|
Seth D, D'Souza El-Guindy NB, Apte M, Mari M, Dooley S, Neuman M, Haber PS, Kundu GC, Darwanto A, de Villiers WJ, Vonlaufen A, Xu Z, Phillips P, Yang S, Goldstein D, Pirola RM, Wilson JS, Moles A, Fernández A, Colell A, García-Ruiz C, Fernández-Checa JC, Meyer C, Meindl-Beinker NM. Alcohol, signaling, and ECM turnover. Alcohol Clin Exp Res 2010; 34:4-18. [PMID: 19860812 DOI: 10.1111/j.1530-0277.2009.01060.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2023]
Abstract
Alcohol is recognized as a direct hepatotoxin, but the precise molecular pathways that are important for the initiation and progression of alcohol-induced tissue injury are not completely understood. The current understanding of alcohol toxicity to organs suggests that alcohol initiates injury by generation of oxidative and nonoxidative ethanol metabolites and via translocation of gut-derived endotoxin. These processes lead to cellular injury and stimulation of the inflammatory responses mediated through a variety of molecules. With continuing alcohol abuse, the injury progresses through impairment of tissue regeneration and extracellular matrix (ECM) turnover, leading to fibrogenesis and cirrhosis. Several cell types are involved in this process, the predominant being stellate cells, macrophages, and parenchymal cells. In response to alcohol, growth factors and cytokines activate many signaling cascades that regulate fibrogenesis. This mini-review brings together research focusing on the underlying mechanisms of alcohol-mediated injury in a number of organs. It highlights the various processes and molecules that are likely involved in inflammation, immune modulation, susceptibility to infection, ECM turnover and fibrogenesis in the liver, pancreas, and lung triggered by alcohol abuse.
Collapse
Affiliation(s)
- Devanshi Seth
- Drug Health Services & Centenary Institute, Royal Prince Alfred Hospital, Camperdown, NSW, Australia.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
71
|
Bartel DP, Nakamura I, Roberts LR, Scimè A, Rudnicki MA. MicroRNAs: target recognition and regulatory functions. Cell 2009. [PMID: 23403079 DOI: 10.1016/j] [Citation(s) in RCA: 482] [Impact Index Per Article: 32.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
MicroRNAs (miRNAs) are endogenous approximately 23 nt RNAs that play important gene-regulatory roles in animals and plants by pairing to the mRNAs of protein-coding genes to direct their posttranscriptional repression. This review outlines the current understanding of miRNA target recognition in animals and discusses the widespread impact of miRNAs on both the expression and evolution of protein-coding genes.
Collapse
Affiliation(s)
- David P Bartel
- Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| | | | | | | | | |
Collapse
|