Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
J Hepatol ; 52(3): 407-16, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20129692

ABSTRACT

BACKGROUND & AIMS: Adverse alcohol effects in the liver involve oxidative metabolism, fat deposition and release of fibrogenic mediators, including TGF-beta. The work presents an assessment of liver damaging cross-talk between ethanol and TGF-beta in hepatocytes. METHODS: To investigate TGF-beta effects on hepatocytes, microarray analyses were performed and validated by qRT-PCR, Western blot analysis and immunohistochemistry. The cellular state was determined by assessing lactate dehydrogenase, cellular glutathione, reactive oxygen species, lipid peroxidation and neutral lipid deposition. RNA interference was used for gene silencing in vitro. RESULTS: TGF-beta is induced in mouse livers after chronic ethanol insult, enhances ethanol induced oxidative stress and toxicity towards cultured hepatocytes plus induces lipid-, oxidative stress metabolism- and fibrogenesis-gene expression signatures. Interestingly, TGF-beta down-regulates alcohol metabolizing enzyme Adh1 mRNA in cultured hepatocytes and liver tissue from TGF-beta transgenic mice via the ALK5/Smad2/3 signalling branch, with Smad7 as a potent negative regulator. ADH1 deficiency is a determining factor for the increased lipid accumulation and Cyp2E1 dependent toxicity in liver cells upon alcohol challenge. Further, ADH1 expression was decreased during liver damage in an intragastric ethanol infusion mouse model. CONCLUSION: In the presence of ethanol, TGF-beta displays pro-steatotic action in hepatocytes via decreasing ADH1 expression. Low ADH1 levels are correlated with enhanced hepatocyte damage upon chronic alcohol consumption by favoring secondary metabolic pathways.


Subject(s)
Alcohol Dehydrogenase/metabolism , Chemical and Drug Induced Liver Injury/metabolism , Down-Regulation/physiology , Ethanol/adverse effects , Hepatocytes/metabolism , Transforming Growth Factor beta/metabolism , Animals , Cells, Cultured , Chemical and Drug Induced Liver Injury/pathology , Disease Models, Animal , Hepatocytes/pathology , Lipid Metabolism/physiology , Lipid Peroxidation/physiology , Male , Mice , Mice, Inbred C57BL , Oxidative Stress/physiology , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/physiology , Smad7 Protein/metabolism
2.
Blood ; 115(13): 2657-65, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20040761

ABSTRACT

Hepcidin is the master regulatory hormone of systemic iron metabolism. Hepcidin deficiency causes common iron overload syndromes whereas its overexpression is responsible for microcytic anemias. Hepcidin transcription is activated by the bone morphogenetic protein (BMP) and the inflammatory JAK-STAT pathways, whereas comparatively little is known about how hepcidin expression is inhibited. By using high-throughput siRNA screening we identified SMAD7 as a potent hepcidin suppressor. SMAD7 is an inhibitory SMAD protein that mediates a negative feedback loop to both transforming growth factor-beta and BMP signaling and that recently was shown to be coregulated with hepcidin via SMAD4 in response to altered iron availability in vivo. We show that SMAD7 is coregulated with hepcidin by BMPs in primary murine hepatocytes and that SMAD7 overexpression completely abolishes hepcidin activation by BMPs and transforming growth factor-beta. We identify a distinct SMAD regulatory motif (GTCAAGAC) within the hepcidin promoter involved in SMAD7-dependent hepcidin suppression, demonstrating that SMAD7 does not simply antagonize the previously reported hemojuvelin/BMP-responsive elements. This work identifies a potent inhibitory factor for hepcidin expression and uncovers a negative feedback pathway for hepcidin regulation, providing insight into a mechanism how hepcidin expression may be limited to avoid iron deficiency.


Subject(s)
Antimicrobial Cationic Peptides/biosynthesis , Hepatocytes/metabolism , Iron/metabolism , Smad7 Protein/physiology , Animals , Antimicrobial Cationic Peptides/genetics , Bone Morphogenetic Proteins/pharmacology , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor/drug effects , Cell Line, Tumor/metabolism , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Down-Regulation , Feedback, Physiological , Hepatocytes/drug effects , Hepcidins , Humans , Interleukin-6/pharmacology , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic/genetics , RNA Interference , RNA, Small Interfering/pharmacology , Recombinant Fusion Proteins/physiology , Signal Transduction/drug effects , Signal Transduction/physiology , Smad7 Protein/antagonists & inhibitors , Smad7 Protein/chemistry , Smad7 Protein/genetics , Transforming Growth Factor beta/pharmacology
3.
Alcohol Clin Exp Res ; 33(10): 1647-55, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19645734

ABSTRACT

Chronic alcohol abuse is an important cause of morbidity and mortality throughout the world. Liver damage due to chronic alcohol intoxication initially leads to accumulation of lipids within the liver and with ongoing exposure this condition of steatosis may first progress to an inflammatory stage which leads the way for fibrogenesis and finally cirrhosis of the liver. While the earlier stages of the disease are considered reversible, cirrhotic destruction of the liver architecture beyond certain limits causes irreversible damage of the organ and often represents the basis for cancer development. This review will summarize current knowledge about the molecular mechanisms underlying the different stages of alcoholic liver disease (ALD). Recent observations have led to the identification of new molecular mechanisms and mediators of ALD. For example, plasminogen activator inhibitor 1 was shown to play a central role for steatosis, the anti-inflammatory adipokine, adiponectin profoundly regulates liver macrophage function and excessive hepatic deposition of iron is caused by chronic ethanol intoxication and increases the risk of hepatocellular carcinoma development.


Subject(s)
Liver Diseases, Alcoholic/pathology , Animals , Disease Progression , Fatty Liver, Alcoholic/pathology , Humans , Liver Cirrhosis/pathology , Liver Neoplasms/pathology
4.
Growth Factors ; 27(5): 289-99, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19639490

ABSTRACT

Nerve growth factor (NGF), a survival factor for neurons enforces pain by sensitizing nociceptors. Also in the pancreas, NGF was associated with pain and it can stimulate the proliferation of pancreatic cancer cells. Hepatic stellate cells (HSC) respond to NGF with apoptosis. Transforming growth factor (TGF)-beta, one of the strongest pro-fibrogenic activators of pancreatic stellate cells (PSC) induced NGF and its two receptors in an immortalized human cell line (ihPSC) and primary rat PSC (prPSC) as determined by RT-PCR, western blot, and immunofluorescence. In contrast to HSC, PSC expressed both NGF receptors, although p75(NTR) expression was weak in prPSC. In contrast to ihPSC TGF-beta activated both Smad signaling cascades in prPSC. NGF secretion was diminished by the activin-like kinase (ALK)-5 inhibitor SB431542, indicating the predominant role of ALK5 in activating the NGF system in PSC. While NGF did not affect proliferation or survival of PSC it induced expression of Inhibitor of Differentiation-1. We conclude that under conditions of upregulated TGF-beta, like fibrosis, NGF levels will also increase in PSC which might contribute to pancreatic wound healing responses.


Subject(s)
Nerve Growth Factor/metabolism , Pancreas/cytology , Protein Serine-Threonine Kinases/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/pharmacology , Animals , Cell Differentiation/drug effects , Cell Line, Tumor , Cells, Cultured , Gene Expression Regulation , Humans , Inhibitor of Differentiation Protein 1/genetics , Inhibitor of Differentiation Protein 1/metabolism , Male , Nerve Growth Factor/drug effects , Pancreas/metabolism , Pancreas/pathology , Protein Serine-Threonine Kinases/genetics , Rats , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/genetics , Signal Transduction , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
5.
Gastroenterology ; 135(2): 642-59, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18602923

ABSTRACT

BACKGROUND & AIMS: The profibrogenic role of transforming growth factor (TGF)-beta in liver has mostly been attributed to hepatic stellate cell activation and excess matrix synthesis. Hepatocytes are believed to contribute to increased rates of apoptosis. METHODS: Primary hepatocyte outgrowths and AML12 cells were used as an in vitro model to detect TGF-beta effects on the cellular phenotype and expression profile. Furthermore, a transgenic mouse model was used to determine the outcome of hepatocyte-specific Smad7 expression on fibrogenesis following CCl(4)-dependent damage. Samples from patients with chronic liver diseases were assessed for (partial) epithelial-to-mesenchymal transition (EMT) in hepatocytes. RESULTS: In primary cell cultures and in vivo, the majority of hepatocytes survive despite activated TGF-beta signaling. These cells display phenotypic changes and express proteins characteristic for (partial) EMT and fibrogenesis. Experimental expression of Smad7 in hepatocytes of mice attenuated TGF-beta signaling and EMT, resulted in less accumulation of interstitial collagens, and improved CCl(4)-provoked liver damage and fibrosis scores compared with controls. CONCLUSIONS: The data indicate that hepatocytes undergo TGF-beta-dependent EMT-like phenotypic changes and actively participate in fibrogenesis. Furthermore, ablation of TGF-beta signaling specifically in this cell type is sufficient to blunt the fibrogenic response.


Subject(s)
Cell Transdifferentiation , Hepatocytes/metabolism , Liver Cirrhosis/prevention & control , Smad7 Protein/metabolism , Transforming Growth Factor beta/metabolism , Animals , Apoptosis , Carbon Tetrachloride , Cell Line , Cell Survival , Cells, Cultured , Collagen/metabolism , Disease Models, Animal , Gene Expression Profiling/methods , Hepatitis B/complications , Hepatitis B/metabolism , Hepatitis B/pathology , Hepatocytes/pathology , Humans , Liver Cirrhosis/etiology , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Phenotype , Schistosomiasis/complications , Schistosomiasis/metabolism , Schistosomiasis/pathology , Smad7 Protein/genetics , Time Factors
6.
Hepatology ; 46(4): 1257-70, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17657819

ABSTRACT

UNLABELLED: Connective tissue growth factor (CTGF) is important for transforming growth factor-beta (TGF-beta)-induced liver fibrogenesis. Hepatic stellate cells have been recognized as its major cellular source in the liver. Here we demonstrate the induction of CTGF expression in hepatocytes of damaged livers and identify a molecular mechanism responsible for it. CTGF expression was found by immunohistochemistry in bile duct epithelial cells, hepatic stellate cells, and hepatocytes in fibrotic liver tissue from patients with chronic hepatitis B infection. Similarly, CTGF expression was induced in hepatocytes of carbon tetrachloride-treated mice. CTGF expression and secretion were detected spontaneously in a medium of hepatocytes after 3 days of culture, which was enhanced by stimulation with TGF-beta. TGF-beta-induced CTGF expression was mediated through the activin receptor-like kinase 5 (ALK5)/Smad3 pathway, whereas activin receptor-like kinase 1 activation antagonized this effect. CTGF expression in the liver tissue of TGF-beta transgenic mice correlated with serum TGF-beta levels. Smad7 overexpression in cultured hepatocytes abrogated TGF-beta-dependent and intrinsic CTGF expression, indicating that TGF-beta signaling was required. In line with these data, hepatocyte-specific transgenic Smad7 reduced CTGF expression in carbon tetrachloride-treated animals, whereas in Smad7 knockout mice, it was enhanced. Furthermore, an interferon gamma treatment of patients with chronic hepatitis B virus infection induced Smad7 expression in hepatocytes, leading to decreased CTGF expression and fibrogenesis. CONCLUSION: Our data provide evidence for the profibrogenic activity of TGF-beta directed to hepatocytes and mediated via the up-regulation of CTGF. We identify ALK5-dependent Smad3 signaling as the responsible pathway inducing CTGF expression, which can be hindered by an activated activin receptor-like kinase 1 pathway and completely inhibited by TGF-beta antagonist Smad7.


Subject(s)
Hepatocytes/metabolism , Immediate-Early Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/metabolism , Animals , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Cells, Cultured , Connective Tissue Growth Factor , Disease Models, Animal , Female , Hepatocytes/drug effects , Hepatocytes/pathology , Humans , Interferon-gamma/pharmacology , Interferon-gamma/therapeutic use , Liver Cirrhosis/drug therapy , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptor, Transforming Growth Factor-beta Type I , Signal Transduction/physiology , Smad3 Protein/metabolism , Smad7 Protein/metabolism
7.
J Pediatr Gastroenterol Nutr ; 44(1): 71-6, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17204957

ABSTRACT

OBJECTIVES: Except for polyadipate, which is used as an alternative in polyvinylchloride (PVC) feeding tubes, diethylhexylphthalate (DEHP) is the plasticizer used almost exclusively in PVC medical products. A clear response to the chronic intake of DEHP has been shown in several organ systems from different species. In the present study, we compared the extraction of DEHP and polyadipate from PVC gastric tubes. METHODS: An in vivo setting was simulated. We used 5 cm of PVC nasogastric tubes containing DEHP or polyadipate, which were incubated with gastric juice for 1 week and a feeding solution for 4 weeks. The leakage of these plasticizers was measured daily in the gastric juice group and weekly in the feeding solution group. The amount of plasticizer extracted was compared between groups and days. RESULTS: In the feeding solution group, the extraction of DEHP ranged from 200 to 542 microg after 1 week and from 660 to 1700 microg after 4 weeks. The extraction of polyadipate was 10 times lower than that of DEHP. In the gastric juice group, extracted DEHP ranged from 635 to 1043 microg, whereas the extraction of polyadipate was 100 times lower. CONCLUSIONS: Within 1 week, the extraction of DEHP from a 5-cm PVC tube reaches up to 1 mg. Extrapolated, this represents an in vivo load of up to 4 mg. The load accumulated by a newborn in an intensive care unit can therefore easily reach several milligrams of DEHP per day. Polyadipate nasogastric tubes may therefore be an alternative and help to reduce the daily load of DEHP.


Subject(s)
Diethylhexyl Phthalate/analysis , Enteral Nutrition , Gastric Juice/chemistry , Intubation, Gastrointestinal , Polyvinyl Chloride , Adipates/chemistry , Adolescent , Child , Child, Preschool , Humans , Infant , Infant, Newborn , Plasticizers , Polymers/chemistry
8.
J Cell Mol Med ; 10(4): 922-32, 2006.
Article in English | MEDLINE | ID: mdl-17125595

ABSTRACT

BACKGROUND/AIMS: Profibrogenic TGF-beta signaling in hepatic stellate cells is modulated during transdifferentiation. Strategies to abrogate TGF-beta effects provide promising antifibrotic results, however, in vivo data regarding Smad activation during fibrogenesis are scarce. METHODS: Here, liver fibrosis was assessed subsequent to bile duct ligation by determining liver enzymes in serum and collagen deposition in liver tissue. Activated hepatic stellate cells were identified by immunohistochemistry and immunoblots for alpha smooth muscle actin. Cellular localization of Smad3 and Smad7 proteins was demonstrated by immunohistochemistry. RTPCR for Smad4 and Smad7 was conducted with total RNA and Northern blot analysis for Smad7 with mRNA. Whole liver lysates were prepared to detect Smad2/3/4 and phospho- Smad2/3 by Western blotting. RESULTS: Cholestasis induces TGF-beta signaling via Smad3 in vivo, whereas Smad2 phosphorylation was only marginally increased. Smad4 expression levels were unchanged. Smad7 expression was continuously increasing with duration of cholestasis. Hepatocytes of fibrotic lesions exhibited nuclear staining Smad3. In contrast to this, Smad7 expression was localized to activated hepatic stellate cells. CONCLUSIONS: Hepatocytes of damaged liver tissue display increased TGF-beta signaling via Smad3. Further, negative feedback regulation of TGF-beta signaling by increased Smad7 expression in activated hepatic stellate cells occurs, however does not interfere with fibrogenesis.


Subject(s)
Cholestasis/complications , Liver Cirrhosis/pathology , Smad3 Protein/physiology , Smad7 Protein/biosynthesis , Animals , Bile Ducts/pathology , Immunohistochemistry , Ligation , Liver/enzymology , Liver/pathology , Liver Cirrhosis/etiology , Male , Rats , Rats, Sprague-Dawley , Signal Transduction , Smad3 Protein/metabolism , Smad4 Protein/biosynthesis , Transforming Growth Factor beta/physiology
9.
Hepatology ; 43(5): 1032-41, 2006 May.
Article in English | MEDLINE | ID: mdl-16628634

ABSTRACT

Transforming growth factor (TGF)-beta is critically involved in the activation of hepatic stellate cells (HSCs) that occurs during the process of liver damage, for example, by alcohol, hepatotoxic viruses, or aflatoxins. Overexpression of the TGF-beta antagonist Smad7 inhibits transdifferentiation and arrests HSCs in a quiescent stage. Additionally, bile duct ligation (BDL)-induced fibrosis is ameliorated by introducing adenoviruses expressing Smad7 with down-regulated collagen and alpha-smooth muscle actin (alpha-SMA) expression. The aim of this study was to further characterize the molecular details of TGF-beta pathways that control the transdifferentiation process. In an attempt to elucidate TGF-beta target genes responsible for fibrogenesis, an analysis of Smad7-dependent mRNA expression profiles in HSCs was performed, resulting in the identification of the inhibitor of differentiation 1 (Id1) gene. Ectopic Smad7 expression in HSCs strongly reduced Id1 mRNA and protein expression. Conversely, Id1 overexpression in HSCs enhanced cell activation and circumvented Smad7-dependent inhibition of transdifferentiation. Moreover, knock-down of Id1 in HSCs interfered with alpha-SMA fiber formation, indicating a pivotal role of Id1 for fibrogenesis. Treatment of HSCs with TGF-beta1 led to increased Id1 protein expression, which was not directly mediated by the ALK5/Smad2/3, but the ALK1/Smad1 pathway. In vivo, Id1 expression and Smad1 phosphorylation were co-induced during fibrogenesis. In conclusion, Id1 is identified as TGF-beta/ALK1/Smad1 target gene in HSCs and represents a critical mediator of transdifferentiation that might be involved in hepatic fibrogenesis. Supplementary material for this article can be found on the HEPATOLOGY website (http://interscience.wiley.com/jpages/0270-9139/suppmat/index.html).


Subject(s)
Inhibitor of Differentiation Protein 1/physiology , Liver/cytology , Transforming Growth Factor beta/physiology , Animals , Cell Differentiation , Male , Rats , Rats, Sprague-Dawley , Smad7 Protein/physiology
10.
Alcohol Clin Exp Res ; 29(11 Suppl): 121S-131S, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16344596

ABSTRACT

Permanent alcohol abuse may lead to chronic liver injury with deleterious sequelae such as liver cirrhosis and hepatocellular carcinoma. Mechanisms of fibrogenesis encompass recruitment of inflammatory cells at the site of injury and cytokine mediated activation of hepatic stellate cells (HSC) with accumulation of interstitial collagens. HSC transdifferentiation and accompanying apoptosis result in destruction of liver architecture and are therefore key steps of disease progression. TGF-beta represents the main profibrogenic cytokine in liver fibrosis and other fibroproliferative disorders by inducing extracellular matrix deposition as part of the wound healing response. In parallel, TGF-beta triggers hepatocytes that are strongly responsive for this cytokine, to undergo apoptosis, thereby providing space for HSC proliferation and generation of a collagenous matrix. Anti TGF-beta approaches were established and successfully utilized for the treatment of experimental fibrogenesis. Dominant negative TGF-beta receptors (TbetaR), generated by fusing the Fc domain of human IgG and the N-terminal (extracellular) fragment of TbetaRII (Fc:TbetaRII) were applied to suppress fibrosis. Similarly TGF-beta binding proteins like decorin, antagonistic cytokines such as bone morphogenetic protein-7, hepatocyte growth factor, IL-10, or IFN-gamma were as efficient as camostat mesilate, a protease inhibitor that possibly abrogated proteolytic activation of TGF-beta. Further, our group recently overexpressed Smad7 in bile duct ligation induced liver fibrosis and achieved efficient inhibition of intracellular TGF-beta signaling, thereby counteracting profibrogenic effects in cultured HSC and in vivo. A direct link between the effect of alcohol and TGF-beta exists through reactive oxygen species that are generated in liver cells by alcohol metabolism and represent activators of TGF-beta signaling. Thus, soluble TbetaRII expression reduced experimental fibrogenesis in vitro and in vivo partially by decreasing intracellular ROS and inhibiting NADH oxidase. Approaches that specifically target profibrogenic TGF-beta signaling are promising to treat alcoholic liver disease in the future. However, to ensure safety for the patients to be treated, approaches with strong specificity need to be established. Therefore, it is essential to delineate the profibrogenic actions of TGF-beta and the influence of alcohol abuse in molecular detail.


Subject(s)
Liver Cirrhosis, Alcoholic/drug therapy , Liver Cirrhosis, Experimental/drug therapy , Transforming Growth Factor beta/antagonists & inhibitors , Animals , Fibrosis/physiopathology , Hepatocytes/physiology , Humans , Liver/pathology , Liver Cirrhosis, Alcoholic/physiopathology , Signal Transduction , Transforming Growth Factor beta/physiology
11.
Cytokine ; 31(5): 349-57, 2005 Sep 07.
Article in English | MEDLINE | ID: mdl-16039137

ABSTRACT

The platelet-derived growth factor (PDGF) family, which regulates many physiological and pathophysiological processes has recently been enlarged by two new members, the isoforms PDGF-C and -D. Little is known about the expression levels of these new members in hepatic fibrosis. We therefore investigated by quantitative real time PCR (Taqman) the mRNA expression profiles of all four PDGF isoforms in transdifferentiating primary cultured hepatic stellate cells (HSC), an in vitro model system of hepatic fibrogenesis, either with or without stimulation of the cells with PDGF-BB or TGF-beta1. All four isoforms were expressed in HSC transdifferentiating to myofibroblast-like cells (MFB) albeit with different profiles: while PDGF-A mRNA exhibited minor fluctuations only, PDGF-B was rapidly down-regulated. In contrast, both PDGF-C and -D mRNA were strongly induced: PDGF-C up to 5 fold from day 2 to day 8 and PDGF-D up to 8 fold from day 2 to day 5 of culture. Presence of PDGF-DD in activated HSC was confirmed at the protein level by immunocytochemistry. Stimulation of HSC and MFB with PDGF-BB led to down-regulation of the new isoforms, whereas TGF-beta1 upregulated PDGF-A only. We further show that PDGF receptor-beta (PDGFR-beta) mRNA was rapidly upregulated within the first day of culture and was constantly expressed from day 2 on while the expression profile of PDGFR-alpha mRNA was very similar to that of PDGF-A during transdifferentiation. Given the dramatic changes in PDGF-C and -D expression, which may compensate for down-regulation of PDGF-B, we hypothesize that the new PDGF isoforms may fulfil specific functions in hepatic fibrogenesis.


Subject(s)
Liver/cytology , Lymphokines/biosynthesis , Platelet-Derived Growth Factor/biosynthesis , Proto-Oncogene Proteins c-sis/biosynthesis , Receptor, Platelet-Derived Growth Factor alpha/biosynthesis , Receptor, Platelet-Derived Growth Factor beta/biosynthesis , Animals , Cell Differentiation , Cells, Cultured , Cytokines/metabolism , Down-Regulation , Fibroblasts/metabolism , Fibrosis , Immunohistochemistry , Liver/metabolism , Male , Microscopy, Fluorescence , Platelet-Derived Growth Factor/chemistry , Protein Isoforms , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transforming Growth Factor beta/metabolism , Up-Regulation
12.
Growth Factors ; 23(2): 77-85, 2005 Jun.
Article in English | MEDLINE | ID: mdl-16019429

ABSTRACT

Accumulating evidence has identified Thrombospondin (TSP)-1 as important activator of latent TGF-beta. Since little is known about signal transduction pathways regulating TSP expression in liver, we investigated cytokine-mediated upregulation of TSP-1 and TGF-beta1 in primary rat hepatic stellate cells (HSC). PDGF-BB and TNF-a rapidly coinduce mRNA levels of TSP-1 and TGF-beta1. Interestingly, blockade of basal Erk activity by synthetic Erk-binding peptides also leads to strong induction of both mRNA transcripts in non-stimulated cells. We show that PDGF-BB induces TSP-1 and TGF-beta1 via the src kinase pathway whereas TNF-a utilizes the MAPK/Erk pathway. However, especially TSP-1 induction by both cytokines involves a pathway, which depends to a certain extent on PI3 kinase activity. In summary the data illustrate specific pathways activated by PDGF-BB and TNF-a in HSC giving new insights into the tightly controlled mechanisms regulating TSP-1 and TGF-beta1 expression in these cells.


Subject(s)
Gene Expression Regulation, Enzymologic , Hepatocytes/metabolism , Liver/metabolism , Thrombospondin 1/metabolism , Transforming Growth Factor beta/metabolism , Animals , Becaplermin , Blotting, Northern , Blotting, Western , Cell Differentiation , Cell Line , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Humans , MAP Kinase Signaling System , Models, Biological , Peptides/chemistry , Phosphorylation , Platelet-Derived Growth Factor/metabolism , Proto-Oncogene Proteins c-sis , RNA/metabolism , RNA, Messenger/metabolism , Rats , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism , src-Family Kinases/metabolism
13.
Matrix Biol ; 24(3): 198-207, 2005 May.
Article in English | MEDLINE | ID: mdl-15905080

ABSTRACT

Hepatic stellate cells (HSC) cultured on plastic spontaneously transdifferentiate to a myofibroblast-like cell type (MFB). This model system of hepatic fibrogenesis is characterized by phenotypic changes of the cells and increased matrix synthesis. Here, we analyzed if transdifferentiation-dependent induction of ECM components, e.g., collagen type I and thrombospondin-2 (TSP-2), and phenotypic changes are coregulated events and if both processes are mediated via TGF-beta pathway(s). Blocking the TGF-beta-dependent p38 MAPK pathway in HSC with the specific inhibitor SB203580 strongly reduces collagen I and TSP-2 mRNA expression without inhibiting upregulation of the typical MFB-marker, alpha-smooth-muscle actin (alpha-SMA). Similarly, interference with the Smad2/3/4 pathway using dexamethasone also heavily decreased expression of collagen type I and TSP-2 whereas transdifferentiation of HSC to the typical morphology of MFB with loss of fat droplets and increasing alpha-SMA was unchanged. Further, p38 MAPK mediated induction of collagen I and TSP-2 expression by TGF-beta1 was still achieved in the presence of dexamethasone, showing that dexamethasone does not block p38 while it delays Smad2 phosphorylation and antagonizes stimulation of a Smad3/Smad4 dependent TGF-beta reporter construct. Interestingly, in contrast to SB203580 and dexamethasone, overexpression of the TGF-beta antagonist Smad7 reduced ECM expression and simultaneously inhibited morphologic transdifferentiation, indicating that Smad7 fulfills additional features in HSC. In conclusion, our data show that phenotypic changes of transdifferentiating HSC and induction of matrix synthesis are independent processes, the latter being stimulated by both, Smad dependent and MAPK dependent TGF-beta signaling.


Subject(s)
Actins/metabolism , Collagen Type I/metabolism , Liver/cytology , Liver/metabolism , Muscle, Smooth/metabolism , Thrombospondins/metabolism , Transforming Growth Factor beta/metabolism , Animals , Cell Differentiation/physiology , Collagen Type I/antagonists & inhibitors , DNA-Binding Proteins/metabolism , Dexamethasone/pharmacology , Extracellular Matrix/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Glucocorticoids/pharmacology , Humans , MAP Kinase Signaling System/physiology , Male , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley , Smad2 Protein , Thrombospondins/antagonists & inhibitors , Trans-Activators/metabolism , Transforming Growth Factor beta/physiology , Transforming Growth Factor beta1 , p38 Mitogen-Activated Protein Kinases/metabolism
14.
Biochem Biophys Res Commun ; 325(4): 1264-70, 2004 Dec 24.
Article in English | MEDLINE | ID: mdl-15555563

ABSTRACT

Glucocorticoids bound to their receptors transmit information, which regulates numerous physiological and pathophysiological responses, amongst others glucose metabolism, wound healing, inflammation, and stress, either directly as transcription factors by binding DNA elements of target genes or indirectly by protein-protein interactions with other transcription factors. TGF-beta, a key factor in activation of hepatic stellate cells (HSC), induces production of extracellular matrix, this being a prerequisite for the development of liver fibrosis. Glucocorticoids and their receptors may provide a crosstalk with the TGF-beta-Smad signaling pathway by antagonizing TGF-beta effects. We studied the influence of glucocorticoids on the TGF-beta isoform and Smad mRNA expression, TGF-beta secretion, and signaling in activated HSC using gene-specific real-time PCR, ELISA, and transfection techniques. Dexamethasone treatment reduces TGF-beta mRNA transcription in a time-dependent manner. Activated HSC produce TGF-beta and secrete it into the cell culture medium. After dexamethasone treatment, TGF-beta secretion into the medium is reduced dose-dependently but restorable by mifepristone. Further, we found that reduced secretion of endogenous TGF-beta is accompanied by a reduced TGF-beta signal. Additionally, reporter gene analysis after adenoviral infection with a recombinant virus encoding a Smad-binding-element showed that TGF-beta-Smad signaling is significantly down-regulated by dexamethasone in primary HSC and CFSC, a HSC related cell line. Our data suggest that glucocorticoids inhibit TGF-beta expression, prevent TGF-beta from efficient secretion, and finally lead to reduced TGF-beta signaling in primary HSC.


Subject(s)
Adrenal Cortex Hormones/pharmacology , Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Hepatocytes/drug effects , Hepatocytes/metabolism , Signal Transduction/physiology , Transforming Growth Factor beta/metabolism , Animals , Biological Availability , Cells, Cultured , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Male , Rats , Signal Transduction/drug effects , Smad Proteins , Trans-Activators/metabolism
15.
Hepatol Res ; 28(3): 140-145, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15036070

ABSTRACT

Transdifferentiation of hepatic stellate cells (HSC) to collagen producing myofibroblasts (MFB) is a principal event in liver fibrogenesis. In our studies we investigated if tissue transglutaminase (tTG) from these cell types may play a role in liver fibrosis. Separation of cytosol and membrane components showed membrane associated tTG and during transdifferentiation an upregulation of total tTG on mRNA and protein level was found, but no modulation during stimulation with TGF-beta1. In HSC and fully differentiated MFB a significant amount of the total tTG synthesised during transdifferentiation is found to be membrane-associated whereas the remaining portion is cytosol-associated and only very little is found within the extracellular matrix (ECM). The data implicate that tTG in this cell type seems to play an important role in liver fibrogenesis.

16.
Gastroenterology ; 125(1): 178-91, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12851882

ABSTRACT

BACKGROUND & AIMS: Numerous studies implicate transforming growth factor (TGF)-beta signaling in liver fibrogenesis. To perturb the TGF-beta pathway during this process, we overexpressed Smad7, an intracellular antagonist of TGF-beta signaling, in vivo and in primary-cultured hepatic stellate cells (HSCs). METHODS: Ligation of the common bile duct (BDL) was used to induce liver fibrosis in rats. Animals received injections of an adenovirus carrying Smad7 cDNA into the portal vein during surgery and via the tail vein at later stages. The effect of Smad7 on TGF-beta signaling and activation of HSC was further analyzed in primary-cultured cells. RESULTS: Smad7-overexpressing BDL rats displayed reduced collagen and alpha-SMA expression and reduced hydroxyproline content in the liver, when compared with animals administered AdLacZ. Such a beneficial effect was also observed when Smad7 was expressed in animals with established fibrosis. Accordingly, Smad7 arrested transdifferentiation of primary-cultured HSCs. AdSmad7 infected cells remained in a quiescent stage and retained storage of vitamin A droplets. Smad7 expression totally blocked TGF-beta signal transduction, shown by inhibiting Smad2/3 phosphorylation, nuclear translocation of activated Smad complexes, and activation of (CAGA)(9)-MLP-Luc, resulting in decreased collagen I expression. Smad7 also abrogated TGF-beta-dependent proliferation inhibition of HSC. Smad7 did not decrease expression of alpha-SMA, but immunofluorescent staining with anti alpha-SMA antibodies displayed destruction of the fibrillar organization of the actin cytoskeleton. CONCLUSIONS: In summary, gene transfer of Smad7 inhibits experimental fibrogenesis in vivo. Studies with isolated HSC suggest that the underlying mechanisms involve inhibition of TGF-beta signaling and HSC transdifferentiation.


Subject(s)
DNA-Binding Proteins/genetics , Genetic Therapy , Liver Cirrhosis/prevention & control , Liver/cytology , Trans-Activators/genetics , Actins/genetics , Animals , Autocrine Communication , Cell Division , Cells, Cultured , Collagen Type I/biosynthesis , Gene Expression , Liver Cirrhosis/pathology , Mice , Paracrine Communication , Rats , Rats, Sprague-Dawley , Signal Transduction , Smad7 Protein , Transforming Growth Factor beta/metabolism , Transgenes
17.
Growth Factors ; 21(3-4): 121-30, 2003.
Article in English | MEDLINE | ID: mdl-14708940

ABSTRACT

TGF-beta, a profibrogenic cytokine is predominantly secreted as a latent molecule complexed with one of the latent TGF-beta binding proteins (LTBP). Due to the proposed functions of LTBP-1 and -3 in regulating TGF-beta-bioavailability and -activity, we investigated the effects of PDGF-BB and TGF-beta1 on their expression levels in Cirrhotic fat storing cells (CFSC). CFSC basally express LTBP-1 and -3 and TGF-beta1. LTBP-1 colocalizes with LAP and the cells secrete some active TGF-beta1. Promoter studies showed no strong induction of the LTBP-1 promoters after stimulation, although mRNA and protein levels were increased by PDGF-BB treatment without affecting TGF-beta1 expression. Vice versa, TGF-beta1 treatment did not alter LTBP-1 expression while an autocrine induction was found. Our data indicate that LTBP-1 but not TGF-beta1 is induced by PDGF-BB and that TGF-beta1 autoinduction does not affect the expression of LTBP-beta1. This divergent regulation may represent an important mechanism for modulation of TGF-beta bioavailability.


Subject(s)
Adipocytes/metabolism , Anticoagulants/pharmacology , Carrier Proteins/biosynthesis , Intracellular Signaling Peptides and Proteins , Platelet-Derived Growth Factor/pharmacology , Transforming Growth Factor beta/biosynthesis , Animals , Becaplermin , Blotting, Northern , Blotting, Western , Cell Division , Cell Line , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Fibrosis/metabolism , Hepatocytes/metabolism , Humans , Latent TGF-beta Binding Proteins , Luciferases/metabolism , Promoter Regions, Genetic , Proto-Oncogene Proteins c-sis , RNA, Messenger/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transfection , Transforming Growth Factor beta1
18.
Front Biosci ; 7: d793-807, 2002 Apr 01.
Article in English | MEDLINE | ID: mdl-11897555

ABSTRACT

TGF-beta has multiple profibrogenic but also anti-inflammatory and immunosuppressive effects. The balance of these actions is required for maintaining tissue homeostasis and an aberrant expression of TGF-beta is involved in a number of disease processes in the liver. In addition to its fibrogenic action leading to transdifferentiation of hepatic stellate cells into myofibroblasts, TGF-beta is also an important negative regulator of proliferation and an inducer of apoptosis. The major portion of TGF-beta is secreted as part of an inactive complex and the details of the activation process in liver have not yet been elucidated. The initially striking simplicity of the core TGF-beta/Smad signaling pathways is rapidly giving way to a much more complex view of intracellular signal transduction mechanisms and recent work has demonstrated the importance of cross-talk among different signaling pathways to either specify, enhance, or inhibit TGF-beta responses. The ubiquitous pathophysiologic relevance of TGF-beta suggests its measurement in blood as a diagnostic tool. Other strategies aim at inhibition of TGF-beta1 function or synthesis as a primary target for the development of antifibrotic strategies and recent advances in cell biology have opened several ways to approach the inhibition of TGF-beta action.


Subject(s)
Liver Cirrhosis/etiology , Transforming Growth Factor beta/physiology , Activins/pharmacology , Animals , Apoptosis , Humans , Liver/cytology , Liver/metabolism , Liver Cirrhosis/diagnosis , Liver Cirrhosis/therapy , Models, Biological , Models, Molecular , Rats , Signal Transduction , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/blood , Transforming Growth Factor beta/chemistry , Transforming Growth Factor beta1
SELECTION OF CITATIONS
SEARCH DETAIL
...