Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 21(15)2020 Jul 23.
Article in English | MEDLINE | ID: mdl-32718038

ABSTRACT

The lack of Lipocalin (LCN2) provokes overwhelming endoplasmic reticulum (ER) stress responses in vitro and in acute toxic liver injury models, resulting in hepatocyte apoptosis. LCN2 is an acute phase protein produced in hepatocytes in response to acute liver injuries. In line with these findings we investigated ER stress responses of Lcn2-/- mice in chronic ER stress using a long-term repetitive carbon tetrachloride (CCl4) injection model. We found chronic CCl4 application to enhance ER stress and unfolded protein responses (UPR), including phosphorylation of eukaryotic initiation factor 2α (eIF2α), increased expression of binding immunoglobulin protein (BiP) and glucose-regulated protein 94 (GRP94). IRE1α/TRAF2/JNK signaling enhanced mitochondrial apoptotic pathways, and showed slightly higher in Lcn2-/- mice compared to the wild type counterparts, leading to increased hepatocyte apoptosis well evidenced by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining. Hepatocyte injuries were confirmed by significant high serum alanine transaminase (ALT) levels in CCl4-treated Lcn2-/- mice. Lcn2-/- mice furthermore developed mild hepatic steatosis, supporting our finding that ER stress promotes lipogenesis. In a previous report we demonstrated that the pharmacological agent tunicamycin (TM) induced ER stress through altered protein glycosylation and induced high amounts of C/EBP-homologous protein (CHOP), resulting in hepatocyte apoptosis. We compared TM-induced ER stress in wild type, Lcn2-/-, and Chop null (Chop-/-) primary hepatocytes and found Chop-/- hepatocytes to attenuate ER stress responses and resist ER stress-induced hepatocyte apoptosis through canonical eIF2α/GADD34 signaling, inhibiting protein synthesis. Unexpectedly, in later stages of TM incubation, Chop-/- hepatocytes resumed activation of IRE1α/JNK/c-Jun and p38/ATF2 signaling, leading to late hepatocyte apoptosis. This interesting observation indicates Chop-/- mice to be unable to absolutely prevent all types of liver injury, while LCN2 protects the hepatocytes by maintaining homeostasis under ER stress conditions.


Subject(s)
Apoptosis , Carbon Tetrachloride Poisoning/metabolism , Carbon Tetrachloride/toxicity , Endoplasmic Reticulum Stress , Hepatocytes/metabolism , Lipocalin-2/deficiency , Unfolded Protein Response , Animals , Apoptosis/drug effects , Apoptosis/genetics , Carbon Tetrachloride Poisoning/genetics , Carbon Tetrachloride Poisoning/pathology , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/genetics , Hepatocytes/pathology , Lipocalin-2/metabolism , Lipogenesis/drug effects , Lipogenesis/genetics , Mice , Mice, Knockout , Signal Transduction/drug effects , Signal Transduction/genetics , Unfolded Protein Response/drug effects , Unfolded Protein Response/genetics
2.
Cell Signal ; 55: 90-99, 2019 03.
Article in English | MEDLINE | ID: mdl-30615971

ABSTRACT

Unfolded protein response (UPR) is an adaptive mechanism allowing the endoplasmic reticulum (ER) to react to an accumulation of unfolded proteins in its lumen, also known as ER stress. The UPR is interconnected with inflammation through several pathways such as reactive oxygen species (ROS) production resulting from the protein folding or alternatively, activation of nuclear factor-κB (NF-κB) and c-Jun N-terminal kinase (JNK) via IRE1, or induction of acute phase response (APR). Lipocalin 2 (LCN2) is one of the APR proteins induced under inflammatory conditions and up-regulated during ER stress. Upon incubation of Lcn2-/- and wild type (wt) primary hepatocytes with tunicamycin (TM) or thapsigargin (TG) we found the Lcn2-/- hepatocytes to react with strong UPR to the ER stress, as evidenced by significantly increased levels of Grp94, Bip and Chop mRNA and protein compared to the wt. TM and TG-treated hepatocytes activated p65 NF-κB and JNK, the pathways that respond to stress stimuli and playing a central role in inflammation and apoptosis, respectively. ER stress further activated and cleaved full-length CREBH/CREB3L3, the hepatocyte specific transcription factor to induce systemic inflammatory responses. Upregulation of the C/EBP homologous protein (CHOP) was very prominent in Lcn2-/- hepatocytes and sustained until 48 h, resulting in hepatocyte apoptosis as evidenced by increased cleaved caspase 3. We also explored the UPR of the Lcn2 null mouse livers in acute intoxication and inflammation stages with a single application of lipopolysaccharide (LPS) or carbon tetrachloride (CCl4). The Lcn2 null mice clearly developed stronger UPR in LPS- and CCl4-induced ER stress compared to the wt. Our findings indicate that the upregulation of LCN2 during ER stress-induced inflammatory responses protects hepatocytes from being overwhelmed by UPR upon liver injury.


Subject(s)
Endoplasmic Reticulum Stress/physiology , Hepatocytes/metabolism , Inflammation/metabolism , Lipocalin-2/physiology , Liver , Unfolded Protein Response/physiology , Animals , Hepatocytes/cytology , Liver/cytology , Liver/metabolism , Mice , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism
3.
Cell Signal ; 51: 72-85, 2018 11.
Article in English | MEDLINE | ID: mdl-30044965

ABSTRACT

Portal fibroblasts are mesenchyme-derived fibroblasts surrounding the bile ducts, and activated into portal myofibroblasts (pMF) during cholestatic liver injury. pMF express α-smooth muscle actin (α-SMA) and produce the fibrogenic extracellular matrix (ECM) collagen type I and fibronectin, playing important roles in portal fibrosis. A cholestatic bile duct-ligated (BDL) model is characterized by impaired hepatobiliary excretion of bile, leading to increased bile acid accumulation. Accumulation of bile acids is known to induce endoplasmic reticulum (ER) stress leading to liver damage and cell death. Additionally, a BDL fibrotic model is also associated with upregulation of CCN (CYR61, CTGF and NOV) matricellular proteins and reported to induce ER stress both in vitro and in vivo. To explore the effects of CCN proteins, we used adenovirus-mediated CCN1-4 (Ad-CCN1-4) gene transfers into cultured pMF. Overexpression of CCN proteins leads to protein accumulation in the ER lumen, causing ER stress and unfolded protein response (UPR). We further found ER stress and UPR to mitigate fibrogenesis in pMF by decreased cellular production of fibronectin, collagen type 1 and α-SMA. In this scenario, Tauroursodeoxycholic acid, a pharmaceutical chaperone and ER stress inhibitor, attenuated Ad-CCN1-4 induced pMF apoptosis and restored collagen and fibronectin levels. Since hepatic fibrogenesis is accompanied by ER stress and upregulation of CCN proteins in a BDL, we further evaluated ER stress responses after Ad-CCN1 gene transfer in such a model and found overexpressed CCN1 to enhance the ER stress-associated proteins BiP and CHOP with positive cleaved caspase 3 and 9 staining in periportal nonparenchymal cells. This indicates that these nonparenchymal cells, most likely pMF, have the tendency to undergo apoptosis during later stages of BDL. Ad-CCN1 transduction furthermore sensitized pMF for ER stress and apoptosis. We suggest that CCN proteins are key factors in the fibrotic microenvironment impacting pMF survival during fibrogenesis and pMF apoptosis during fibrosis resolution.


Subject(s)
Bile Ducts, Intrahepatic/metabolism , CCN Intercellular Signaling Proteins/physiology , Cholestasis/metabolism , Endoplasmic Reticulum/metabolism , Myofibroblasts/metabolism , Actins/metabolism , Animals , Apoptosis , Bile Ducts, Intrahepatic/pathology , Caspase 3/metabolism , Caspase 9/metabolism , Cells, Cultured , Cholestasis/pathology , Collagen Type I/metabolism , Endoplasmic Reticulum Stress , Fibronectins/metabolism , Fibrosis , Heat-Shock Proteins/metabolism , Male , Mice , Myofibroblasts/cytology , Rats , Rats, Sprague-Dawley , Signal Transduction , Transcription Factor CHOP/metabolism , Unfolded Protein Response
4.
Front Pharmacol ; 9: 426, 2018.
Article in English | MEDLINE | ID: mdl-29755357

ABSTRACT

Lipocalin 2 (LCN2) is a highly conserved secreted adipokine acting as a serum transport protein for small hydrophobic molecules such as fatty acids and steroids. In addition, LCN2 limits bacterial growth by sequestering iron-containing siderophores and further protects against intestinal inflammation and tumorigenesis associated with alterations in the microbiota. Human LCN2 contains one N-glycosylation site conserved in other species. It was postulated that this post-translational modification could facilitate protein folding, protects from proteolysis, is required for proper trafficking from the Golgi apparatus to the cell surface, and might be relevant for effective secretion. We here show that the homologous nucleoside antibiotic tunicamycin blocks N-linked glycosylation but not secretion of LCN2 in primary murine hepatocytes, derivatives thereof, human lung carcinoma cell line A549, and human prostate cancer cell line PC-3. Moreover, both the glycosylated and the non-glycosylated LCN2 variants are equally targeted to exosomes, demonstrating that this post-translational modification is not necessary for proper trafficking of LCN2 into these membranous extracellular vesicles. Furthermore, a hydrophobic cluster analysis revealed that the N-glycosylation site is embedded in a highly hydrophobic evolutionarily conserved surrounding. In sum, our data indicate that the N-glycosylation of LCN2 is not required for proper secretion and exosome cargo recruitment in different cell types, but might be relevant to increase overall solubility.

5.
Biochim Biophys Acta ; 1863(11): 2604-2612, 2016 11.
Article in English | MEDLINE | ID: mdl-27452908

ABSTRACT

The endoplasmic reticulum (ER) is primarily recognized as the site of synthesis and folding of secreted membrane-bound and certain organelle-targeted proteins. Optimum protein folding requires several factors, including ATP, Ca2+ and an oxidizing environment to allow disulphide-bond formation. ER is highly sensitive to stress that perturb cellular energy levels, the redox state or the Ca2+ concentration. Such stresses reduce the protein folding capacity of the ER, resulting in the accumulation and aggregation of unfolded proteins, a condition referred to as unfolded protein response (UPR). Matricellular proteins of the CCN (CYR61, CTGF, NOV) family play essential roles in extracellular matrix signaling and turnover. They exhibit a similar type of organization and share a closely related primary structure, including 38 conserved cysteine residues. Since CCN1/CYR61 overexpression in hepatic stellate cells (HSC) induces ER stress-related apoptosis, we endeavored to investigate whether the adenovirus mediated gene transfer of other members of CCN proteins incurs ER stress in primary HSC and hepatocytes. We found Ad5-CMV-CCN2, Ad5-CMV-CCN3 and Ad5-CMV-CCN4 to induce ER stress and UPR comparable to Ad5-CMV-CCN1. UPR is a pro-survival response to reduce accumulation of unfolded proteins and restore normal ER functioning. If, however protein aggregation is persistent and the stress cannot be resolved, signaling switches from pro-survival to pro-apoptosis. The observed CCN-induced UPR is relevant in wound healing responses and essential for hepatic tissue repair following liver injury. Adenoviral gene transfer induced massive amounts of matricellular proteins proving to effectively mitigate liver fibrosis if targeted cell specific in HSC and myofibroblasts.


Subject(s)
Adenoviridae/genetics , CCN Intercellular Signaling Proteins/metabolism , Endoplasmic Reticulum Stress , Endoplasmic Reticulum/metabolism , Genetic Vectors , Liver Cirrhosis, Experimental/metabolism , Liver/metabolism , Transduction, Genetic , Transfection/methods , Unfolded Protein Response , Animals , Apoptosis , CCN Intercellular Signaling Proteins/genetics , Cells, Cultured , Cellular Senescence , Endoplasmic Reticulum/pathology , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Hepatocytes/metabolism , Hepatocytes/pathology , Liver/pathology , Liver Cirrhosis, Experimental/etiology , Liver Cirrhosis, Experimental/genetics , Liver Cirrhosis, Experimental/pathology , Male , Mice, Inbred C57BL , Myofibroblasts/metabolism , Myofibroblasts/pathology , Protein Aggregates , Rats, Sprague-Dawley , Signal Transduction
6.
Cell Signal ; 28(1): 34-42, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26515130

ABSTRACT

CCN1/CYR61 is a matricellular protein of the CCN family, comprising six secreted proteins specifically associated with the extracellular matrix (ECM). CCN1 acts as an enhancer of the cutaneous wound healing process by preventing hypertrophic scar formation through induction of myofibroblast senescence. In liver fibrosis, the senescent cells are primarily derived from activated hepatic stellate cells (HSC) that initially proliferate in response to liver damage and are the major source of ECM. We investigate here the possible use of CCN1 as a senescence inducer to attenuate liver fibrogenesis by means of adenoviral gene transfer in primary HSC, myofibroblasts (MFB) and immortalized HSC lines (i.e. LX-2, CFSC-2G). Infection with Ad5-CMV-CCN1 induced large amounts of CCN1 protein in all these cells, resulting in an overload of the endoplasmic reticulum (ER) and in a compensatory unfolded protein response (UPR). The UPR resulted in upregulation of ER chaperones including BIP/Grp78, Grp94 and led to an activation of IRE1α as evidenced by spliced XBP1 mRNA with IRE1α-induced JNK phosphorylation. The UPR arm PERK and eIF2a was phosphorylated, combined with significant CHOP upregulation. Ad5-CMV-CCN1 induced HSC apoptosis that was evident by proteolytic cleavage of caspase-12, caspase-9 and the executor caspase-3 and positive TUNEL stain. Remarkably, Ad5-CMV-CCN1 effectively reduced collagen type I mRNA expression and protein. We conclude that the matricellular protein CCN1 gene transfer induces HSC apoptosis through ER stress and UPR.


Subject(s)
Apoptosis/physiology , Cysteine-Rich Protein 61/metabolism , Endoplasmic Reticulum Stress/physiology , Endoplasmic Reticulum/metabolism , Hepatic Stellate Cells/metabolism , Unfolded Protein Response/physiology , Animals , Caspase 3/metabolism , Liver Cirrhosis/metabolism , Myofibroblasts/metabolism , Rats, Sprague-Dawley
7.
Cell Signal ; 27(7): 1305-14, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25819339

ABSTRACT

UNLABELLED: Platelet-derived growth factor-D (PDGF-D) is one member of PDGF growth factors and known to signal by binding to and activating its cognate receptor type ß (PDGFR-ß). Beside PDGF-B, PDGF-D is a potent growth factor for stellate cell growth and proliferation and therefore potentiates the extracellular matrix deposition in liver fibrogenesis. We aimed to explore the signaling and molecular mechanisms of PDGF-D in liver fibrogenesis using the primary liver portal myofibroblasts and hepatic stellate cells. Unexpectedly we found PDGF-D to bind to PDGFR-α, thus inducing receptor endocytosis and decreasing the amount of PDGFR-α significantly. PDGF-D activates PDGFR-α specific tyrosine 754 and -1018 phosphorylation and CrkII, the adaptor protein that is specifically recruited by activated PDGFR-α. As a novel finding we could also demonstrate that recombinant PDGFR-α-Fc chimera homodimer is able to bind PDGF-D and thus prevent PDGF-D signaling. PDGF-D does induce individual PDGFR-ß specific tyrosine phosphorylation similar to the PDGF-B. Additionally, PDGF-D enhances extracellular matrix accumulation comparable to the PDGF-B isoform. CONCLUSION: PDGF-D signaling in pMF and HSC is identical to that of PDGF-B by binding to both PDGFR-α and -ß.


Subject(s)
Lymphokines/pharmacology , Platelet-Derived Growth Factor/pharmacology , Proto-Oncogene Proteins c-sis/pharmacology , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Signal Transduction/drug effects , Animals , Becaplermin , Cells, Cultured , Down-Regulation/drug effects , Endocytosis/drug effects , Hepatic Stellate Cells/cytology , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Male , Myofibroblasts/cytology , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Protein Binding , Rats , Rats, Sprague-Dawley , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor beta/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Tissue Inhibitor of Metalloproteinase-1/metabolism , Up-Regulation/drug effects
8.
Biochem Biophys Res Commun ; 458(3): 700-706, 2015 Mar 13.
Article in English | MEDLINE | ID: mdl-25686493

ABSTRACT

The inflammasomes are cytoplasmic multiprotein complexes that are responsible for activation of inflammatory reactions. In principle, there are four individual inflammasome branches (NLRP1, NLRP3, NLRC4/NALP4, and AIM2) that mediate the cleavage and activation of Caspase-1 and IL-1ß that in turn lead to a complex network of cellular reactions initiating local and systemic inflammatory reactions. We have recently shown that NLRP3 expression is virtually absent in primary cultured hepatocytes and that in vitro the stimulation of hepatocytes with lipopolysaccharides results in strong activation of NLRP3 expression. We here demonstrate that this activation can be blocked by the NF-κB activation inhibitor QNZ or by infection with an adenoviral expression vector constitutively expressing a superrepressor of NF-κB. We show that QNZ blocks NF-κB-dependent expression of TNF-α, IL-1ß and NLRP3. Likewise, the superrepressor of NF-κB prevents expression of NLRP3 and significantly reduces expression of inflammatory marker genes in liver cells. In a primary murine hepatoma cells, the concomitant depletion of NEMO and Caspase-8 resulted in a significant suppression of NLRP3 expression after Lipopolysaccharide challenge. Moreover, we demonstrate that a 1.3-kbp fragment located in close proximity of the most upstream transcriptional start site of the human NLRP3 gene that harbours one putative octamer NF-κB binding site renders LPS sensitivity in reporter gene assay. We conclude that NF-κB signalling is a necessary prerequisite for proper activation of the NLRP3 inflammasome in primary hepatocytes.


Subject(s)
Carrier Proteins/genetics , Hepatocytes/immunology , Inflammasomes/genetics , NF-kappa B/immunology , Animals , Carrier Proteins/immunology , Caspase 8/immunology , Cells, Cultured , Gene Expression Regulation/drug effects , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Inflammasomes/immunology , Intracellular Signaling Peptides and Proteins/immunology , Lipopolysaccharides/immunology , Male , Mice , Mice, Inbred C57BL , NF-kappa B/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein
9.
Biochim Biophys Acta ; 1843(5): 902-14, 2014 May.
Article in English | MEDLINE | ID: mdl-24487063

ABSTRACT

UNLABELLED: Cysteine-rich protein 61 (CCN1/CYR61) is a CCN (CYR61, CTGF (connective tissue growth factor), and NOV (Nephroblastoma overexpressed gene)) family matricellular protein comprising six secreted CCN proteins in mammals. CCN1/CYR61 expression is associated with inflammation and injury repair. Recent studies show that CCN1/CYR61 limits fibrosis in models of cutaneous wound healing by inducing cellular senescence in myofibroblasts of the granulation tissue which thereby transforms into an extracellular matrix-degrading phenotype. We here investigate CCN1/CYR61 expression in primary profibrogenic liver cells (i.e., hepatic stellate cells and periportal myofibroblasts) and found an increase of CCN1/CYR61 expression during early activation of hepatic stellate cells that declines in fully transdifferentiated myofibroblasts. By contrast, CCN1/CYR61 levels found in primary parenchymal liver cells (i.e., hepatocytes) were relatively low compared to the levels exhibited in hepatic stellate cells and portal myofibroblasts. In models of ongoing liver fibrogenesis, elevated levels of CCN1/CYR61 were particularly noticed during early periods of insult, while expression declined during prolonged phases of fibrogenesis. We generated an adenovirus type 5 encoding CCN1/CYR61 (i.e., Ad5-CMV-CCN1/CYR61) and overexpressed CCN1/CYR61 in primary portal myofibroblasts. Interestingly, overexpressed CCN1/CYR61 significantly inhibited production of collagen type I at both mRNA and protein levels as evidenced by quantitative real-time polymerase chain reaction, Western blot and immunocytochemistry. CCN1/CYR61 further induces production of reactive oxygen species (ROS) leading to dose-dependent cellular senescence and apoptosis. Additionally, we demonstrate that CCN1/CYR61 attenuates TGF-ß signaling by scavenging TGF-ß thereby mitigating in vivo liver fibrogenesis in a bile duct ligation model. CONCLUSION: In line with dermal fibrosis and scar formation, CCN1/CYR61 is involved in liver injury repair and tissue remodeling. CCN1/CYR61 gene transfer into extracellular matrix-producing liver cells is therefore potentially beneficial in liver fibrotic therapy.


Subject(s)
Apoptosis , Cellular Senescence , Cysteine-Rich Protein 61/physiology , Myofibroblasts/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Animals , Male , Rats , Rats, Sprague-Dawley
10.
Biochim Biophys Acta ; 1832(5): 660-73, 2013 May.
Article in English | MEDLINE | ID: mdl-23376114

ABSTRACT

Lipocalin-2 is expressed under pernicious conditions such as intoxication, infection, inflammation and other forms of cellular stress. Experimental liver injury induces rapid and sustained LCN2 production by injured hepatocytes. However, the precise biological function of LCN2 in liver is still unknown. In this study, LCN2(-/-) mice were exposed to short term application of CCl4, lipopolysaccharide and Concanavalin A, or subjected to bile duct ligation. Subsequent injuries were assessed by liver function analysis, qRT-PCR for chemokine and cytokine expression, liver tissue Western blot, histology and TUNEL assay. Serum LCN2 levels from patients suffering from liver disease were assessed and evaluated. Acute CCl4 intoxication showed increased liver damage in LCN2(-/-) mice indicated by higher levels of aminotransferases, and increased expression of inflammatory cytokines and chemokines such as IL-1ß, IL-6, TNF-α and MCP-1/CCL2, resulting in sustained activation of STAT1, STAT3 and JNK pathways. Hepatocytes of LCN2(-/-) mice showed lipid droplet accumulation and increased apoptosis. Hepatocyte apoptosis was confirmed in the Concanavalin A and lipopolysaccharide models. In chronic models (4weeks bile duct ligation or 8weeks CCl4 application), LCN2(-/-) mice showed slightly increased fibrosis compared to controls. Interestingly, serum LCN2 levels in diseased human livers were significantly higher compared to controls, but no differences were observed between cirrhotic and non-cirrhotic patients. Upregulation of LCN2 is a reliable indicator of liver damage and has significant hepato-protective effect in acute liver injury. LCN2 levels provide no correlation to the degree of liver fibrosis but show significant positive correlation to inflammation instead.


Subject(s)
Acute-Phase Proteins/metabolism , Homeostasis , Lipocalins/metabolism , Liver Diseases/metabolism , Liver/metabolism , Oncogene Proteins/metabolism , Acute Disease , Acute-Phase Proteins/genetics , Animals , Apoptosis , Bile Ducts/surgery , Blotting, Western , Carbon Tetrachloride/toxicity , Chemokines/genetics , Chemokines/metabolism , Concanavalin A/toxicity , Cytokines/genetics , Cytokines/metabolism , Gene Expression , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Immunohistochemistry , Ligation/adverse effects , Lipocalin-2 , Lipocalins/blood , Lipocalins/genetics , Lipopolysaccharides/toxicity , Liver/pathology , Liver Diseases/etiology , Liver Diseases/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Oncogene Proteins/genetics , Proto-Oncogene Proteins/blood , Reverse Transcriptase Polymerase Chain Reaction
11.
Liver Int ; 32(9): 1342-53, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22698069

ABSTRACT

BACKGROUND AND AIMS: CCN3/NOV, a matricellular protein of the CYR61-CTGF-NOV (CCN) family, comprises six secreted proteins that associate specifically with the extracellular matrix. CCN proteins lack specific high-affinity receptors; instead, they regulate crucial biological processes, such as fibrosis, by signalling via integrins and proteoglycans. Recent studies have linked overexpression of CCN3/NOV to mitigate kidney fibrosis. This study aims to investigate CCN3/NOV overexpression in liver fibrogenesis in vivo. METHODS: The biological efficacy of adenoviral expressed CCN3/NOV directed under transcriptional control of the constitutively active Cytomegalovirus promoter (Ad-NOV) was analysed in a bile duct ligation model and in cultured primary hepatocytes. RESULTS AND CONCLUSIONS: Even though Ad-NOV gene transfer in a 3-week bile duct ligation mouse model showed the expected high levels of CCN3/NOV in both mRNA and protein, it failed to reduce liver fibrogenesis, but instead enhanced hepatocyte apoptosis. Furthermore, overexpressed CCN3/NOV in cultured primary hepatocytes resulted in decreased levels of CCN2/CTGF, the profibrotic marker protein in liver fibrosis. Both Ad-NOV and Ad-CTGF induced reactive oxygen species production, enhanced p38 and JNK activation. Therefore, we conclude that CCN3/NOV overexpression in vivo is insufficient to mitigate liver fibrogenesis because of the induction of hepatocyte injury and apoptosis.


Subject(s)
Adenoviridae/genetics , Apoptosis/physiology , Hepatocytes/pathology , Liver Cirrhosis/pathology , Nephroblastoma Overexpressed Protein/genetics , Animals , Cells, Cultured , Cholestasis, Extrahepatic/complications , Cholestasis, Extrahepatic/genetics , Cholestasis, Extrahepatic/pathology , Common Bile Duct/surgery , Disease Models, Animal , Disease Progression , Gene Expression , Gene Transfer Techniques , Genetic Vectors/genetics , Hepatocytes/metabolism , Ligation , Liver Cirrhosis/etiology , Liver Cirrhosis/genetics , Mice , Mice, Inbred C57BL , Nephroblastoma Overexpressed Protein/metabolism , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism
12.
J Cell Commun Signal ; 6(1): 11-25, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21748432

ABSTRACT

Nephroblastoma overexpressed gene encodes a matricellular protein (CCN3/NOV) of the CCN family, comprising CCN1 (CYR61), CCN2 (CTGF), CCN4 (WISP-1), CCN5 (WISP-2), and CCN6 (WISP-3). CCN proteins are involved in the regulation of mitosis, adhesion, apoptosis, extracellular matrix production, growth arrest and migration in multiple cell types. Compared to CCN2/CTGF, known as a profibrotic protein, the biological role of CCN3/NOV in liver fibrosis remains obscure. In this study we showed ccn3/nov mRNA to increase dramatically following hepatic stellate cell activation, reaching peak levels in fully transdifferentiated myofibroblasts. In models of experimental hepatic fibrosis, CCN3/NOV increased significantly at the mRNA and protein levels. CCN3/NOV was found mainly in non-parenchymal cells along the areas of tissue damage and repair. In the bile-duct ligation model, CCN3/NOV was localized mainly along portal tracts, while the repeated application of carbon tetrachloride resulted in CCN3/NOV expression mainly in the centrilobular areas. In contrast to CCN2/CTGF, the profibrotic cytokines platelet-derived growth factor-B and -D as well as transforming growth factor-ß suppressed CCN3/NOV expression. In vitro, CCN3/NOV siRNA attenuated migration in the cirrhotic fat storing cell line CFSC well in line with in vivo findings that various types of cells expressing CCN3/NOV migrate into the area of tissue damage and regeneration. The suppression of CCN3/NOV enhanced expression of profibrotic marker proteins, such as α-smooth muscle actin, collagen type I, fibronectin, CCN2/CTGF and TIMP-1 in primary rat hepatic stellate cells and in CFSC. We further found that adenoviral overexpression of CCN2/CTGF suppressed CCN3/NOV expression, while the overexpression of CCN3/NOV as well as the suppression of CCN3/NOV by targeting siRNAs both resulted in enhanced CCN2/CTGF expression. These results indicate the complexity of CCN actions that are far beyond the classic Yin/Yang interplay.

13.
Biochem Biophys Res Commun ; 345(4): 1526-35, 2006 Jul 14.
Article in English | MEDLINE | ID: mdl-16735029

ABSTRACT

Transforming growth factor-beta (TGF-beta) is a cytokine implicated in differentiation of smooth muscle cells and other mesenchymal-derived cells. During hepatic fibrogenesis, TGF-beta has a pivotal role in the initiation, promotion, and progression of transdifferentiation of hepatic stellate cells into myofibroblasts that play a central role in the synthesis of extracellular matrix components. Both, smooth muscle and activated hepatic stellate cells, express smooth muscle alpha-actin, the calponin-related protein SM22alpha, and CSRP2 encoding the cysteine- and glycine-rich LIM domain protein 2 (CRP2). The aim of the present study was to determine whether the expression of CSRP2 is influenced by TGF-beta. Stimulation as well as sequestering experiments demonstrated that TGF-beta markedly influences CSRP2 gene activity. Inhibition experiments using the ALK5 inhibitor SB-431542 further reveal that the transcriptional stimulation of the CSRP2 gene is mediated via the ALK5/Smad2/Smad3 signalling pathway. By use of bisulfite genomic analysis of CpG islands within the 5' regulatory regions we could exclude methylation-associated silencing, previously found to be responsible for the transcriptional inactivity of CSRP2 in a variety of human cancer cells and in a multistage carcinogenesis model, as a cause for CSRP2 inactivity in hepatocytes or fully transdifferentiated myofibroblasts.


Subject(s)
Liver/drug effects , Muscle Proteins/metabolism , Muscle, Smooth, Vascular/drug effects , Nuclear Proteins/metabolism , Transforming Growth Factor beta/pharmacology , Activin Receptors, Type I/antagonists & inhibitors , Activin Receptors, Type I/physiology , Animals , Base Sequence , Benzamides/pharmacology , Blotting, Northern , Blotting, Western , Cell Line , Cells, Cultured , DNA Methylation , Dioxoles/pharmacology , Gene Expression/drug effects , Immunohistochemistry , LIM Domain Proteins , Liver/cytology , Liver/metabolism , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Male , Molecular Sequence Data , Muscle Proteins/genetics , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Nuclear Proteins/genetics , Promoter Regions, Genetic/genetics , Protein Serine-Threonine Kinases , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/physiology , Signal Transduction/drug effects , Transforming Growth Factor beta1 , Up-Regulation/genetics
14.
J Hepatol ; 44(5): 910-7, 2006 May.
Article in English | MEDLINE | ID: mdl-16169118

ABSTRACT

BACKGROUND/AIMS: Isolated rat hepatic stellate cells (HSC) are taken as a valuable in vitro model to study hepatic fibrogenesis, biotransformation of pharmaceutics, gene expression, transcription factors controlling HSC behaviour, and for the establishment of long-term cultures. Consequently, methods for the isolation and maintenance of HSC cultures are well documented. However, there is ongoing controversial discussion directed on the existence and cellular origin of different HSC subpopulations. Thus, there is a continuing need for developing methods allowing the exchange of HSC isolates between different laboratories. A practical solution to this problem is cryopreservation and banking of HSC. METHODS: We here describe for the first time the successful establishment of a methodology for long-term cryopreservation and recovery of primary, non-activated HSC from rats. We have optimised critical factors for HSC-banking including prefreeze processing, freezing rate, freezing medium, final cooling temperature, and thawing conditions. We found that DMSO gave far superior attachment and viability on thawing than other cryoprotectants. The viability and cellular characteristics of thawed cells was comparatively analysed by light- and electron microscopic analysis, proliferation assay, Oil Red O-staining, apoptosis testing, and evaluation of marker proteins for fibrogenic activities. RESULTS: In summary, our data reveal no significant differences in the biochemical and cellular properties between cryopreserved/thawed and freshly isolated HSC. CONCLUSIONS: According to these results, we suggest that cryoprotected HSC retain functional integrity thereby allowing banking and comfortable exchange of these cells between different laboratories.


Subject(s)
Cell Survival , Cryopreservation/methods , Liver/cytology , Animals , Apoptosis , Azo Compounds , Cell Differentiation , Cells, Cultured , Cold Temperature , Coloring Agents , Cryoprotective Agents/pharmacology , Dimethyl Sulfoxide/pharmacology , Fibroblasts/cytology , Fluoresceins , Male , Propidium , Rats , Rats, Sprague-Dawley , Trypan Blue
15.
Liver Int ; 24(1): 69-79, 2004 Feb.
Article in English | MEDLINE | ID: mdl-15102003

ABSTRACT

BACKGROUND/AIMS: The activation of hepatic stellate cells (HSC) and their transdifferentiation into myofibroblasts (MFB) is the key step for development of liver fibrosis. Over the past several years, significant progress has been made in the understanding of the critical pathways involved incells undergoing activation. Cellular activation in the course of transdifferentiation involves, among other biochemical modifications, functionally relevant changes in the control of gene expression. These include the up-regulation of transcription factors, different extracellular matrix proteins, cell adhesion molecules, smooth muscle specific genes, and proteins involved in matrix remodelling, or cytoskeletal organization. The corresponding regulatory elements of these genes have afforded us the opportunity to express transgenes with antifibrotic potential in a cell type- and/or transdifferentiation-dependent manner. METHODS: In the present study, we have tested several promoters for their ability to mediate cell-specific expression, including those for CSRP2, SM22alpha, and TIMP-1 (CSRP2, gene encoding the LIM domain protein CRP2; SM22alpha, smooth muscle-specific gene encoding a 22-kDa protein; TIMP-1, gene encoding the tissue inhibitor of metalloproteinases-1), which in liver are specifically expressed in HSC or become strongly activated during the acute remodelling into MFB. We constructed adenoviral reporter vectors in which relevant portions of the promoters were fused to the green fluorescent protein. RESULTS AND CONCLUSION: Our experiments demonstrate that each of these promoters is sufficient to achieve strong or partially selective expression in vitro but none is able to direct a specific or inducible expression of transgenes in HSC/MFB in vivo.


Subject(s)
Gene Expression/genetics , Liver/cytology , Microfilament Proteins/genetics , Muscle Proteins/genetics , Nuclear Proteins/genetics , Tissue Inhibitor of Metalloproteinase-1/genetics , Animals , Cell Differentiation , Cell Line , LIM Domain Proteins , Liver/physiopathology , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Male , Models, Animal , Promoter Regions, Genetic/genetics , Rats , Rats, Sprague-Dawley , Transgenes
16.
Biochem Biophys Res Commun ; 316(4): 1107-15, 2004 Apr 16.
Article in English | MEDLINE | ID: mdl-15044099

ABSTRACT

Liver fibrosis is the result from a relative imbalance between synthesis and degradation of matrix proteins. Following liver injury of any etiology, hepatic stellate cells undergo a response known as activation, which is the transition of quiescent cells into proliferative, fibrogenic, and contractile myofibroblasts. Upon this cellular transdifferentiation the effector cell becomes the major source of fibrillar and non-fibrillar matrix proteins resulting in excessive scar formation and cirrhosis, the end stage of fibrosis. Concomitant with progressive liver fibrosis, the tissue inhibitor of metalloproteinases-1 (TIMP-1) is strongly activated in hepatic stellate cells. We have developed a recombinant replication-defective adenovirus in which the TIMP-1 promoter is coupled to the herpes simplex virus thymidine kinase gene rendering activated hepatic stellate cells susceptible to ganciclovir. This novel targeted suicide gene approach was validated in a culture model considered to reflect an accelerated time course of the cellular and molecular events that occur during liver fibrosis. We demonstrate that transfer of the suicide gene to culture-activated hepatic stellate cells results in a strong expression of the respective transgene as assessed by Northern blot and Western blot analyses. The enzyme catalyzed the proper conversion of its prodrug subsequently initiating programmed cell death as estimated by caspase-3 assay and Annexin V-Fluos staining. Altogether, these results indicate that induction of programmed cell death is a promising approach to eliminate fibrogenic HSC.


Subject(s)
Apoptosis/genetics , Gene Expression Regulation/genetics , Gene Targeting/methods , Hepatocytes/metabolism , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Tissue Inhibitor of Metalloproteinase-1/genetics , Tissue Inhibitor of Metalloproteinase-1/metabolism , Animals , Cell Differentiation , Cells, Cultured , Ganciclovir/metabolism , Gene Silencing , Hepatocytes/classification , Hepatocytes/pathology , Liver Cirrhosis/pathology , Male , Rats , Rats, Sprague-Dawley , Thymidine Kinase/genetics , Thymidine Kinase/metabolism
17.
Cell Growth Differ ; 13(6): 265-73, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12114216

ABSTRACT

Liver fibrosis occurs as a consequence of the transdifferentiationof hepatic stellate cells into myofibroblasts and is associated with an increased expression and activation of transforming growth factor (TGF)-beta1. This pluripotent, profibrogenic cytokine stimulates matrix synthesis and decreases matrix degradation, resulting in fibrosis. Thus, blockade of synthesis or sequestering of mature TGF-beta1 is a primary target for the development of antifibrotic approaches. The purpose of this study was to investigate whether the administration of adenoviruses constitutively expressing an antisense mRNA complementary to the 3' coding sequence of TGF-beta1 is able to suppress the synthesis of TGF-beta1 in culture-activated hepatic stellate cells. We demonstrate that the adenoviral vehicle directs high-level expression of the transgene and proved that the transduced antisense is biologically active by immunoprecipitation, Western blot, quantitative TGF-beta1 ELISA, and cell proliferation assays. Additionally, the biological function of the transgene was confirmed by analysis of differential activity of TGF-beta1-responsive genes using cell ELISA, Northern blotting, and by microarray technology, respectively. Furthermore, we examined the effects of that transgene on the expression of TGF-beta2, TGF-beta3, collagen type alpha1(I), latent transforming growth factor binding protein 1, types I and II TGF-beta receptors, and alpha-smooth muscle actin. Our results indicate that the administration of antisense mRNA offers a feasible approach to block autocrine TGF-beta1 signaling in hepatic stellate cells and may be useful and applicable in future to the treatment of fibrosis in chronic liver diseases.


Subject(s)
Adenoviridae/genetics , Intracellular Signaling Peptides and Proteins , Liver/drug effects , RNA, Antisense/pharmacology , RNA, Complementary , Transforming Growth Factor beta/genetics , Activin Receptors, Type I/metabolism , Animals , Blotting, Northern , Blotting, Southern , Carrier Proteins/metabolism , Cell Differentiation/drug effects , Cells, Cultured , Collagen Type I/metabolism , Enzyme-Linked Immunosorbent Assay , Latent TGF-beta Binding Proteins , Liver/cytology , Liver/metabolism , Liver Cirrhosis/prevention & control , Male , Open Reading Frames , Protein Serine-Threonine Kinases , Rats , Rats, Sprague-Dawley , Receptor, Transforming Growth Factor-beta Type I , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1 , Transforming Growth Factor beta2 , Transforming Growth Factor beta3
SELECTION OF CITATIONS
SEARCH DETAIL
...