Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters

Publication year range
2.
Hepatology ; 55(2): 540-52, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21953779

ABSTRACT

UNLABELLED: The role of progenitor cells in liver repair and fibrosis has been extensively described, but their purification remains a challenge, hampering their characterization and use in regenerative medicine. To address this issue, we developed an easy and reproducible liver progenitor cell (LPC) isolation strategy based on aldehyde dehydrogenase (ALDH) activity, a common feature shared by many progenitor cells. We demonstrate that a subset of nonparenchymal mouse liver cells displays high levels of ALDH activity, allowing the isolation of these cells by fluorescence-activated cell sorting. Immunocytochemistry and qPCR analyses on freshly isolated ALDH(+) cells reveal an enrichment in cells expressing liver stem cell markers such as EpCAM, CK19, CD133, and Sox9. In culture, the ALDH(+) population can give rise to functional hepatocyte-like cells as illustrated by albumin and urea secretion and cytochrome P450 activity. ALDH1A1 expression can be detected in canals of Hering and bile duct epithelial cells and is increased on liver injury. Finally, we showed that the isolation and differentiation toward hepatocyte-like cells of LPCs with high ALDH activity is also successfully applicable to human liver samples. CONCLUSION: High ALDH activity is a feature of LPCs that can be taken advantage of to isolate these cells from untreated mouse as well as human liver tissues. This novel protocol is practically relevant, because it provides an easy and nontoxic method to isolate liver stem cells from normal tissue for potential therapeutic purposes.


Subject(s)
Aldehyde Dehydrogenase/metabolism , Liver/cytology , Stem Cells/cytology , AC133 Antigen , Aldehyde Dehydrogenase 1 Family , Animals , Antigens, CD/metabolism , Antigens, Neoplasm/metabolism , Cell Adhesion Molecules/metabolism , Cell Differentiation , Epithelial Cell Adhesion Molecule , Glycoproteins/metabolism , Hepatocytes/cytology , Humans , Keratin-19/metabolism , Mice , Peptides/metabolism , Retinal Dehydrogenase , SOX9 Transcription Factor/metabolism , Stem Cells/enzymology
3.
J Biol Chem ; 285(40): 31011-23, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20610391

ABSTRACT

Accumulation of fat in the liver increases the risk to develop fibrosis and cirrhosis and is associated with development of the metabolic syndrome. Here, to identify genes or gene pathways that may underlie the genetic susceptibility to fat accumulation in liver, we studied A/J and C57Bl/6 mice that are resistant and sensitive to diet-induced hepatosteatosis and obesity, respectively. We performed comparative transcriptomic and lipidomic analysis of the livers of both strains of mice fed a high fat diet for 2, 10, and 30 days. We found that resistance to steatosis in A/J mice was associated with the following: (i) a coordinated up-regulation of 10 genes controlling peroxisome biogenesis and ß-oxidation; (ii) an increased expression of the elongase Elovl5 and desaturases Fads1 and Fads2. In agreement with these observations, peroxisomal ß-oxidation was increased in livers of A/J mice, and lipidomic analysis showed increased concentrations of long chain fatty acid-containing triglycerides, arachidonic acid-containing lysophosphatidylcholine, and 2-arachidonylglycerol, a cannabinoid receptor agonist. We found that the anti-inflammatory CB2 receptor was the main hepatic cannabinoid receptor, which was highly expressed in Kupffer cells. We further found that A/J mice had a lower pro-inflammatory state as determined by lower plasma levels and IL-1ß and granulocyte-CSF and reduced hepatic expression of their mRNAs, which were found only in Kupffer cells. This suggests that increased 2-arachidonylglycerol production may limit Kupffer cell activity. Collectively, our data suggest that genetic variations in the expression of peroxisomal ß-oxidation genes and of genes controlling the production of an anti-inflammatory lipid may underlie the differential susceptibility to diet-induced hepatic steatosis and pro-inflammatory state.


Subject(s)
Dietary Fats/adverse effects , Fatty Liver/metabolism , Gene Expression Regulation/drug effects , Lipid Metabolism/drug effects , Microsomes, Liver/metabolism , Peroxisomes/metabolism , Animals , Dietary Fats/pharmacology , Fatty Liver/chemically induced , Fatty Liver/genetics , Fatty Liver/pathology , Gene Expression Regulation/genetics , Granulocyte Colony-Stimulating Factor/biosynthesis , Granulocyte Colony-Stimulating Factor/genetics , Inflammation/chemically induced , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Interleukin-1beta/biosynthesis , Interleukin-1beta/genetics , Kupffer Cells/metabolism , Kupffer Cells/pathology , Lipid Metabolism/genetics , Lipids/biosynthesis , Lipids/genetics , Male , Mice , Microsomes, Liver/pathology , Peroxisomes/genetics , Peroxisomes/pathology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptor, Cannabinoid, CB2/biosynthesis , Receptor, Cannabinoid, CB2/genetics , Species Specificity , Transcription, Genetic/drug effects , Transcription, Genetic/genetics
4.
J Hepatol ; 54(1): 132-41, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20932596

ABSTRACT

BACKGROUND & AIMS: Upon liver injury, hepatic stellate cells (HSCs) undergo dramatic morphological and functional changes including migration and contraction. In the present study, we investigated the role of myosin II isoforms in the development of the contractile phenotype of mouse HSCs, which are considered therapeutic targets to decrease portal hypertension and fibrosis. METHODS: We characterized the expression of myosin IIA and IIB in primary mouse HSCs and addressed their function by gene knock-down using isoform-specific siRNAs. RESULTS: We found that myosin IIA and IIB are differentially expressed and localized and have clearly different functions in HSCs. Myosin IIA is mainly located in the subcortical area of quiescent HSCs and at α-SMA-containing stress fibres after activation, while myosin IIB is located in the cytoplasm and at the edge of protrusions of quiescent HSCs, at stress fibres of activated cells, and at the leading edge of lamellipodia. Knock-down of myosin IIA in HSCs influences cell size and shape, results in the disruption of stress fibres and in a decrease of focal adhesions, and inhibits contractility and intra-cellular Ca(2+) release but increases cell migration. Myosin IIB contributes to the extension of lamellipodia and cell spreading but has no direct role in stress fibres and focal adhesion formation, contraction, or intra-cellular Ca(2+) signalling. CONCLUSIONS: In mouse HSCs, myosin IIA and IIB clearly fulfil distinct roles. Our results provide an insight into the contractile machinery of HSCs, that could be important in the search for new molecules to treat portal hypertension.


Subject(s)
Hepatic Stellate Cells/physiology , Nonmuscle Myosin Type IIA/physiology , Nonmuscle Myosin Type IIB/physiology , Animals , Calcium Signaling/drug effects , Cell Movement/physiology , Endoplasmic Reticulum/physiology , Endothelin-1/pharmacology , Focal Adhesions/physiology , Gene Knockdown Techniques , Hepatic Stellate Cells/cytology , Hepatic Stellate Cells/drug effects , In Vitro Techniques , Mice , Mice, Inbred BALB C , Molecular Motor Proteins/antagonists & inhibitors , Molecular Motor Proteins/genetics , Molecular Motor Proteins/physiology , Nonmuscle Myosin Type IIA/antagonists & inhibitors , Nonmuscle Myosin Type IIA/genetics , Nonmuscle Myosin Type IIB/antagonists & inhibitors , Nonmuscle Myosin Type IIB/genetics , Pseudopodia/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Signal Transduction , Stress Fibers/physiology , Vinculin/metabolism
5.
Hepatology ; 51(2): 603-14, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19957378

ABSTRACT

UNLABELLED: Hepatic stellate cell (HSC) activation is a pivotal step in the pathogenesis of liver fibrosis. The clarification of this transdifferentiation process is therefore important for the development of effective therapies for fibrosis. We analyzed the effect of a histone deacetylase inhibitor, valproic acid (VPA), on mouse HSC transdifferentiation in vitro and in vivo. The exposure of freshly isolated mouse HSCs to 2.5 mM VPA led to increased histone H4 acetylation and inhibited cell proliferation. Expression of stellate cell activation markers analyzed by quantitative polymerase chain reaction and western blotting revealed that treatment with VPA inhibited the induction of activation markers such as Acta2, Lox, Spp1, and Myh11. Treatment of mice with VPA decreased collagen deposition and in vivo activation of stellate cells in the livers of CCl(4)-treated mice. Class I histone deacetylase silencing through RNA interference in mouse HSCs only partially mimicked treatment with VPA. CONCLUSION: Chronic administration of VPA results in a marked decrease in stellate cell activation both in vitro and in vivo. We hypothesize that the VPA effect results partially from class I histone deacetylase inhibition, but that also non-histone deacetylase class I VPA targets are involved in the stellate cell activation process.


Subject(s)
Cell Transdifferentiation/drug effects , Enzyme Inhibitors/administration & dosage , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/physiology , Histone Deacetylase 1/antagonists & inhibitors , Valproic Acid/administration & dosage , Animals , Cells, Cultured , Enzyme Inhibitors/pharmacology , Male , Mice , Mice, Inbred BALB C , Valproic Acid/pharmacology
6.
J Hepatol ; 52(3): 389-97, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20133001

ABSTRACT

BACKGROUND & AIMS: Advanced glycation end products are known to play an important role in the metabolic syndrome and were recently suggested to contribute to liver fibrosis development. However, little is known about the effect of advanced glycation end products on hepatic stellate cells, the major contributors to liver fibrosis development. We therefore studied the effect of advanced glycation end products on reactive oxygen species generation, a main feature for the activation hepatic stellate cells. METHODS: Three different types of advanced glycation end products were generated by BSA incubation with different substrates. The presence of advanced glycation end product receptors was examined by RTq-PCR, immunofluorescence and western blotting. Reactive oxygen species production was measured using DCFH-DA. RESULTS: Hepatic stellate cells express five advanced glycation end product receptors: Galectin-3, CD36, SR-AI, SR-BI and RAGE. All receptors, except SR-BI, showed up-regulation during HSC activation. All three advanced glycation end product types induced reactive oxygen species generation. DPI and NSC, a NADPH oxidase and a Rac1 inhibitor respectively, inhibited reactive oxygen species production. Rottlerin, a molecule often used as a PKCdelta inhibitor, also abrogated reactive oxygen species production. SiRNA mediated knockdown of p47(phox), Rac1 and PKCdelta decreased reactive oxygen species production induced by advanced glycation end products, establishing a role for these proteins in reactive oxygen species induction. CONCLUSIONS: The demonstration of advanced glycation end product-induced reactive oxygen species generation in hepatic stellate cells unveils a potential new route through which advanced glycation end products induce liver fibrosis in the metabolic syndrome.


Subject(s)
Glycation End Products, Advanced/pharmacology , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , NADPH Oxidases/metabolism , Reactive Oxygen Species/metabolism , Animals , Cells, Cultured , Hepatic Stellate Cells/cytology , Male , Mice , Models, Animal , Neuropeptides/metabolism , Protein Kinase C-delta/metabolism , RNA, Small Interfering/pharmacology , Receptor for Advanced Glycation End Products , Receptors, Immunologic/metabolism , Signal Transduction/drug effects , rac GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein
7.
J Hepatol ; 52(1): 117-29, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19913937

ABSTRACT

Many chronic liver diseases can lead to hepatic dysfunction with organ failure. At present, orthotopic liver transplantation represents the benchmark therapy of terminal liver disease. However this practice is limited by shortage of donor grafts, the need for lifelong immunosuppression and very demanding state-of-the-art surgery. For this reason, new therapies have been developed to restore liver function, primarily in the form of hepatocyte transplantation and artificial liver support devices. While already offered in very specialized centers, both of these modalities still remain experimental. Recently, liver progenitor cells have shown great promise for cell therapy, and consequently they have attracted a lot of attention as an alternative or supportive tool for liver transplantation. These liver progenitor cells are quiescent in the healthy liver and become activated in certain liver diseases in which the regenerative capacity of mature hepatocytes and/or cholangiocytes is impaired. Although reports describing liver progenitor cells are numerous, they have not led to a consensus on the identity of the liver progenitor cell. In this review, we will discuss some of the characteristics of these cells and the different ways that have been used to obtain these from rodents. We will also highlight the challenges that researchers are facing in their quest to identify and use liver progenitor cells.


Subject(s)
Hepatocytes/transplantation , Liver Diseases/therapy , Stem Cells , Animals , Cell- and Tissue-Based Therapy , Humans , Liver Regeneration , Mice , Rats , Stem Cell Transplantation , Transplantation, Heterologous
8.
Hepatology ; 49(5): 1664-72, 2009 May.
Article in English | MEDLINE | ID: mdl-19205034

ABSTRACT

UNLABELLED: Peripheral CD8 T-cell tolerance can be generated outside lymphatic tissue in the liver, but the course of events leading to tolerogenic interaction of hepatic cell populations with circulating T-cells remain largely undefined. Here we demonstrate that preferential uptake of systemically circulating antigen by murine liver sinusoidal endothelial cells (LSECs), and not by other antigen-presenting cells in the liver or spleen, leads to cross-presentation on major histocompatibility complex (MHC) I molecules, which causes rapid antigen-specific naïve CD8 T-cell retention in the liver but not in other organs. Using bone-marrow chimeras and a novel transgenic mouse model (Tie2-H-2K(b) mice) with endothelial cell-specific MHC I expression, we provide evidence that cross-presentation by organ-resident and radiation-resistant LSECs in vivo was both essential and sufficient to cause antigen-specific retention of naïve CD8 T-cells under noninflammatory conditions. This was followed by sustained CD8 T-cell proliferation and expansion in vivo, but ultimately led to the development of T-cell tolerance. CONCLUSION: Our results show that cross-presentation of circulating antigens by LSECs caused antigen-specific retention of naïve CD8 T-cells and identify antigen-specific T-cell adhesion as the first step in the induction of T-cell tolerance.


Subject(s)
Antigen Presentation , CD8-Positive T-Lymphocytes/immunology , Cross-Priming , Endothelial Cells/immunology , Liver/immunology , Animals , Antigens/metabolism , Cell Migration Inhibition , Cells, Cultured , Endothelial Cells/metabolism , Immune Tolerance , Mice , Mice, Inbred C57BL , Mice, Transgenic , Ovalbumin/immunology
9.
Hepatology ; 49(2): 646-57, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19177575

ABSTRACT

UNLABELLED: Immune dysregulations in alcoholic liver diseases are still unclear, especially regarding alcoholic hepatitis inflammatory burst. Interleukin-17 (IL-17) is known to enhance neutrophil recruitment. We studied the IL-17 pathway in alcoholic cirrhosis and alcoholic hepatitis. Patients with alcoholic liver disease were compared with patients with chronic hepatitis C virus (HCV) infection or autoimmune liver disease and with healthy controls. IL-17 plasma levels and peripheral blood mononuclear cell secretion were assessed by enzyme-linked immunosorbent assay (ELISA) and T cell phenotype by flow cytometry. IL-17 staining and co-staining with CD3 and myeloperoxidase were performed on liver biopsy specimens. IL-17 receptor expression was studied on liver biopsies and in human hepatic stellate cells as well as their response to recombinant IL-17 by chemotaxis assays. IL-17 plasma levels were dramatically increased in alcoholic liver disease patients. Peripheral blood mononuclear cells of patients with alcoholic liver disease produced higher amounts of IL-17, and their CD4(+) T lymphocytes disclosed an IL-17-secreting phenotype. In the liver, IL-17-secreting cells contributed to inflammatory infiltrates in alcoholic cirrhosis, and alcoholic hepatitis foci disclosed many IL-17(+) cells, including T lymphocytes and neutrophils. In alcoholic liver disease, liver IL-17(+) cells infiltrates correlated to model for end-stage liver disease score, and in alcoholic hepatitis to modified discriminant function. IL-17 receptor was expressed in alcoholic liver disease by hepatic stellate cells, and these cells recruited neutrophils after IL-17 stimulation in a dose-dependent manner through IL-8 and growth related oncogen alpha (GRO-alpha) secretion in vitro. CONCLUSION: Human alcoholic liver disease is characterized by the activation of the IL-17 pathway. In alcoholic hepatitis, liver infiltration with IL-17-secreting cell infiltrates is a key feature that might contribute to liver neutrophil recruitment. (Clinical trials number NCT00610597).


Subject(s)
Interleukin-17/blood , Liver Cirrhosis, Alcoholic/physiopathology , Alanine Transaminase/blood , Aspartate Aminotransferases/blood , C-Reactive Protein/metabolism , Female , Hepatic Stellate Cells/cytology , Hepatic Stellate Cells/physiology , Hepatitis C, Chronic/blood , Hepatitis C, Chronic/pathology , Hepatitis C, Chronic/physiopathology , Hepatitis, Alcoholic/blood , Hepatitis, Alcoholic/physiopathology , Humans , Interleukin-17/metabolism , Liver Cirrhosis/blood , Liver Cirrhosis/pathology , Liver Cirrhosis/physiopathology , Liver Cirrhosis, Alcoholic/blood , Liver Cirrhosis, Alcoholic/pathology , Male , Neutrophils/physiology , Reference Values
10.
J Hepatol ; 51(1): 187-211, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19457566

ABSTRACT

Controlling both growth and differentiation of stem cells and their differentiated somatic progeny is a challenge in numerous fields, from preclinical drug development to clinical therapy. Recently, new insights into the underlying molecular mechanisms have unveiled key regulatory roles of epigenetic marks driving cellular pluripotency, differentiation and self-renewal/proliferation. Indeed, the transcription of genes, governing cell-fate decisions during development and maintenance of a cell's differentiated status in adult life, critically depends on the chromatin accessibility of transcription factors to genomic regulatory and coding regions. In this review, we discuss the epigenetic control of (liver-specific) gene-transcription and the intricate interplay between chromatin modulation, including histone (de)acetylation and DNA (de)methylation, and liver-enriched transcription factors. Special attention is paid to their role in directing hepatic differentiation of primary hepatocytes and stem cells in vitro.


Subject(s)
Epigenesis, Genetic/physiology , Hepatocytes/cytology , Liver/metabolism , Stem Cells/cytology , Transcription, Genetic , Animals , Cell Differentiation , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/physiology , DNA Methylation , Embryonic Stem Cells/cytology , Gene Expression Regulation , Histone Deacetylase Inhibitors , Histone Deacetylases/classification , Histone Deacetylases/physiology , Humans , Liver/cytology , Multipotent Stem Cells/cytology , Pluripotent Stem Cells/cytology , Signal Transduction
11.
Histochem Cell Biol ; 131(3): 313-25, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19052772

ABSTRACT

Hepatic stellate cells (HSCs) have important roles in the pathogenesis of liver fibrosis and cirrhosis. As response to chronic injury HSCs are activated and change from quiescent into myofibroblast-like cells. Several HSC-specific markers have been described in rat or mouse models. The aim of our work was to identify the best marker(s) for human HSCs. To this end we used the automated high throughput NexES IHC staining device (Ventana Medical Systems) to incubate sections under standardized conditions. Formalin fixed paraffin embedded (FFPE) normal and diseased human livers were studied. With immunohistochemistry we examined the expression of synemin, desmin, vimentin, vinculin, neurotrophin-3 (NT-3), alpha-smooth muscle actin (alpha-SMA), cellular retinol-binding protein-1 (CRBP-1), glial fibrillary acidic protein (GFAP), cysteine- and glycine-rich protein 2 (CRP2), and cytoglobin/stellate cell activation-associated protein (cygb/STAP). This is the first study in which a series of HSC markers is compared on serial FFPE human tissues. CRBP-1 clearly stains lobular HSCs without reacting with smooth muscle cells (SMCs) and shows variable cholangiocyte positivity. Vinculin has a similar staining pattern as CRBP-1 but additionally stains SMCs, and (myo)fibroblasts. In conclusion, we therefore propose to use CRBP-1 and/or vinculin to stain HSCs in human liver tissues.


Subject(s)
Hepatic Stellate Cells/chemistry , Retinol-Binding Proteins, Cellular/analysis , Vinculin/analysis , Biomarkers/analysis , Fibroblasts/chemistry , Humans , Immunohistochemistry , Liver/cytology , Liver Diseases/pathology , Myocytes, Smooth Muscle/chemistry , Paraffin Embedding
12.
Hepatology ; 48(2): 624-34, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18627004

ABSTRACT

UNLABELLED: Hepatic stellate cells (HSCs) survive and proliferate in the chronically injured liver. ATP-binding cassette (ABC) transporters play a crucial role in cell viability by transporting toxic metabolites or xenobiotics out of the cell. ABC transporter expression in HSCs and its relevance to cell viability and/or activation have not been reported so far. The aim of this study was to investigate the expression, regulation, and function of multidrug resistance-associated protein (Mrp)-type and multidrug resistance protein (Mdr)-type ABC transporters in activated rat HSCs. Rat HSCs were exposed to cytokines or oxidative stress. ABC transporter expression was determined by quantitative polymerase chain reaction and immunohistochemistry. HSCs were exposed to the Mdr inhibitors verapamil and PSC-833 and the Mrp inhibitor MK571. Mdr and Mrp transporter function was evaluated with flow cytometry. Apoptosis was determined by activated caspase-3 and acridine orange staining, and necrosis was determined by Sytox green nuclear staining. An in vivo model of carbon tetrachloride (CCl(4))-induced liver fibrosis was used. With respect to hepatocytes, activated HSCs expressed high levels of Mrp1 and comparable levels of Mrp3, Mrp4, Mdr1a, and Mdr1b but not the hepatocyte-specific transporters bile salt export pump, Mrp2, and Mrp6. Mrp1 protein staining correlated with desmin staining in livers from CCl(4)-treated rats. Mrp1 expression increased upon activation of HSCs. Cytokines induced Mdr1b expression only. Oxidative stress was not a major regulator of Mdr and Mrp transporter expression. Activated HSCs became necrotic when exposed to the Mrp inhibitors. CONCLUSION: Activated HSCs contain relatively high levels of Mrp1. Mrp-type transporters are required for the viability of activated HSCs. Mrp-dependent export of endogenous metabolites is important for the survival of activated HSCs in chronic liver diseases.


Subject(s)
Liver/cytology , Liver/physiology , Multidrug Resistance-Associated Proteins/metabolism , ATP Binding Cassette Transporter, Subfamily B/biosynthesis , ATP-Binding Cassette Transporters/classification , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/physiology , Animals , Cell Survival/physiology , Cells, Cultured , Hepatocytes/metabolism , Humans , Interferon-gamma/pharmacology , Interleukin-1beta/pharmacology , Liver/drug effects , Liver Cirrhosis, Experimental/metabolism , Liver Cirrhosis, Experimental/pathology , Male , Oxidative Stress/physiology , RNA, Messenger/metabolism , Rats , Rats, Wistar , Tumor Necrosis Factor-alpha/pharmacology , ATP-Binding Cassette Sub-Family B Member 4
13.
Invest New Drugs ; 27(4): 338-46, 2009 Aug.
Article in English | MEDLINE | ID: mdl-18825314

ABSTRACT

The vast majority of preclinical studies of HDAC inhibitors (HDAC-I) focus on the drug-target (cancer) cell interaction, whereas little attention is paid to the effects on non-target healthy cells, which could provide decisive information to eliminate potential cytotoxic compounds at a very early stage during drug development. In the current study we used cultures of primary rat hepatocytes as a read out system to select for the most potent HDAC-I in the group of structural analogues of an archetypal HDAC-I, namely Trichostatin A. This kind of approach allowed selecting compounds with high biological activity and with no apparent toxicity towards cultured hepatocytes.


Subject(s)
Amides/pharmacology , Hepatocytes/drug effects , Histone Deacetylase Inhibitors , Hydroxamic Acids/pharmacology , Amides/chemistry , Amides/toxicity , Animals , Cells, Cultured , Drug Evaluation, Preclinical , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/toxicity , Hepatocytes/metabolism , Histone Deacetylase 1 , Hydroxamic Acids/chemistry , Hydroxamic Acids/toxicity , Male , Rats , Rats, Sprague-Dawley
14.
Liver Int ; 28(1): 107-16, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18173562

ABSTRACT

BACKGROUND AND AIMS: Chronic liver damage causes hepatic stellate cell (HSC) activation and contraction, leading to intrahepatic microvascular and structural changes. In vitro endothelin-1 (ET-1)-induced contraction of HSCs can be reduced by somatostatin (SST); however, intrahepatic in vivo effects have never been studied. METHODS: Sinusoidal diameter was measured by intravital fluorescence microscopy in carbon tetrachloride (CCl(4)) and control mice before and after an intravenous (IV) bolus and after 0, 5, 10 and 15 min of an IV infusion of saline, 8 microg/kg/h SST or 8 microg/kg/h octreotide. RESULTS: The baseline sinusoidal diameter in CCl(4) mice (3.01+/-0.05 microm) was significantly smaller than that in controls (4.37+/-0.06 microm). The sinusoidal diameter increased significantly in both groups after a bolus (27, 16% respectively) and following 5 min of SST IV infusion (28, 14% respectively). The percentage increase was significantly higher in CCl(4) mice as compared with controls. This dilatory effect continued for at least 15 min. SST did not influence the mean arterial blood pressure (MAP) and portal venous inflow. In none of the groups did octreotide or saline have any influence on sinusoidal diameters, MAP and portal venous inflow. CONCLUSIONS: Sinusoidal diameter in cirrhotic mice is significantly smaller than that in controls. SST causes significant sinusoidal dilation following a bolus and for at least 15 min of IV infusion. Octreotide does not have any influence on liver sinusoids. These results demonstrate for the first time the in vivo dilatory effect of SST on liver sinusoids.


Subject(s)
Liver Cirrhosis/drug therapy , Liver/blood supply , Liver/drug effects , Octreotide/pharmacology , Somatostatin/pharmacology , Animals , Mice , Microcirculation/drug effects , Microscopy, Fluorescence , Octreotide/therapeutic use , Somatostatin/therapeutic use
15.
Nephrol Dial Transplant ; 23(11): 3464-71, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18503097

ABSTRACT

BACKGROUND: Cellular retinol-binding protein I (CRBP-I), a member of the intracellular lipid-binding protein (iLBP) superfamily, is a specific marker of quiescent stellate cells in the healthy human liver. In the diseased fibrotic/cirrhotic liver, portal and septal myofibroblasts acquire CRBP-I expression, while activated hepatic stellate cells maintain their CRBP-I expression. Here, we investigate the distribution of CRBP-I in the renal cortex of healthy rats and rats with renal fibrosis. METHODS: Kidneys of healthy and adriamycin-treated rats were studied by immunohistochemistry, using antibodies against CRBP-I, desmin, vimentin and alpha-smooth muscle actin (alpha-SMA). Double stainings were done with immunofluorescence. Western blotting was performed to semi-quantify the expression levels of vimentin, desmin, alpha-SMA and CRBP-I. RESULTS: In the normal rat kidney, the convoluted proximal tubular epithelial cells express CRBP-I; no expression is found in the interstitium, nor in the glomeruli. In the adriamycin-induced fibrotic rat kidney, CRBP-I expression diminishes in the convoluted proximal tubular epithelial cells, whereas peritubular myofibroblasts in the interstitium acquire CRBP-I expression. CONCLUSIONS: In the tubulointerstitial compartment of the adriamycin-induced fibrotic rat kidney, CRBP-I is expressed in a different pattern than in the healthy rat kidney. As the convoluted proximal tubular epithelial cells dedifferentiate during fibrosis, CRBP-I expression decreases. Furthermore, de novo expression of CRBP-I is found in activated myofibroblast-like cells in the interstitium of adriamycin-treated rats. CRBP-I is therefore a useful marker to identify a subpopulation of activated/ myodifferentiated fibroblasts in the rat kidney.


Subject(s)
Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Nephrosclerosis/metabolism , Nephrosclerosis/pathology , Retinol-Binding Proteins, Cellular/metabolism , Actins/metabolism , Animals , Antibiotics, Antineoplastic , Cell Differentiation , Desmin/metabolism , Disease Models, Animal , Down-Regulation , Doxorubicin , Fibrosis , Male , Nephrosclerosis/chemically induced , Rats , Rats, Wistar , Up-Regulation , Vimentin/metabolism
16.
Toxicol Sci ; 91(2): 484-92, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16531468

ABSTRACT

The effects of histone deacetylase inhibitor Trichostatin A (TSA) on connexin (Cx) expression and gap junctional intercellular communication (GJIC) were investigated in primary cultures of adult rat hepatocytes. GJIC was monitored by using the scrape-loading/dye transfer method. Immunoblotting and immunocytochemistry were used to investigate Cx protein levels and localization. Cx gene expression was studied by means of quantitative reverse transcriptase-polymerase chain reaction. TSA increased Cx32 protein levels and affected negatively the Cx26 protein levels. The latter was preferentially located in the cytosol of cultured cells. TSA also promoted the appearance of Cx43 in the nuclear compartment of primary cultured hepatocytes. Overall, this resulted in enhanced GJIC activity. It is important to note that the time of onset of TSA treatment was crucial for the extent of its outcome and that the effects of TSA on Cx protein levels occurred independently of transcriptional changes. TSA differentially affects Cx proteins in primary rat hepatocyte cultures, suggesting distinct regulation and/or distinct roles of the different Cx species in the control of hepatic homeostasis. TSA enhances GJIC between primary cultured rat hepatocytes, an interesting finding supporting its use to further optimize liver-based in vitro models for pharmacotoxicological purposes.


Subject(s)
Connexins/metabolism , Gap Junctions/drug effects , Hepatocytes/drug effects , Histone Deacetylase Inhibitors , Hydroxamic Acids/pharmacology , Acetylation/drug effects , Animals , Cell Communication/drug effects , Cells, Cultured , Connexins/genetics , Gap Junctions/metabolism , Gene Expression Regulation/drug effects , Hepatocytes/metabolism , Histones/metabolism , L-Lactate Dehydrogenase/metabolism , Male , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley
17.
Br J Pharmacol ; 146(1): 77-88, 2005 Sep.
Article in English | MEDLINE | ID: mdl-15980876

ABSTRACT

Previous studies have shown antifibrotic effects of somatostatin. Since hepatic stellate cells (HSC) express somatostatin receptors and play a key role in hepatic fibrogenesis, we investigated the in vitro antifibrotic effect of somatostatin on rat HSC. At day 12 after isolation, cells were exposed to different concentrations of somatostatin (10(-6)-10(-9) mol l(-1)). mRNA expression of collagen types I and III, and of smooth muscle alpha-actin (alpha-SMA) was analysed by Northern blotting. At 10(-9) mol l(-1), somatostatin significantly reduced mRNA expression of collagen I (72.3 +/- 10.7%; 95% confidence interval (95% CI): 45.5-99.0), collagen III (79.0 +/- 4.5%; 95% CI: 67.6-90.4) and alpha-SMA (65.7 +/- 5.9%; 95% CI: 51.1-80.2), as compared to control normalized at 100%. These results were confirmed by quantitative RT-PCR. Cycloheximide experiments indicated that somatostatin has no direct transcriptional effect.Using immunoprecipitation, we demonstrated that somatostatin also decreased de novo synthesis of collagen I (73 +/-10%; 95% CI: 48-98%), collagen III (65 +/- 13%; 95% CI: 33-97%) and alpha-SMA (47 +/- 9%; 95% CI: 25-69%). Remarkably, at higher concentrations, somatostatin did not suppress collagen mRNA expression nor de novo protein synthesis. We ascribe this observation to desensitization of the cells for somatostatin. Cell proliferation, as measured by 5-bromo-2'-deoxyuridine labelling, was not altered by somatostatin. No significant effect on the intermediate and actin cytoskeleton were detected by immunohistochemistry and Western blotting. Our findings imply that in vivo antifibrotic effects of somatostatin could result partially from a direct action of somatostatin on HSC, but other, in vivo effects are probably also involved.


Subject(s)
Collagen Type III/antagonists & inhibitors , Collagen Type I/antagonists & inhibitors , Gene Expression Regulation/drug effects , Liver/drug effects , Somatostatin/pharmacology , Actins/metabolism , Animals , Cell Proliferation/drug effects , Cells, Cultured , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type III/genetics , Collagen Type III/metabolism , Cycloheximide/pharmacology , Liver/cytology , Liver/metabolism , Male , Protein Synthesis Inhibitors/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Wistar
18.
Biochem Pharmacol ; 66(7): 1307-17, 2003 Oct 01.
Article in English | MEDLINE | ID: mdl-14505810

ABSTRACT

Platelet derived growth factor (PDGF) is a key factor in the induction and progression of fibrotic diseases with the activated fibroblast as its target cell. Drug targeting to the PDGF-receptor is explored as a new approach to treat this disease. Therefore, we constructed a macromolecule with affinity for the PDGF-beta receptor by modification of albumin with a small peptide that recognises this PDGF-beta receptor. The binding of the peptide-modified albumin (pPB-HSA) to the PDGF-beta receptor was confirmed in competition studies with PDGF-BB using NIH/3T3-fibroblasts and activated hepatic stellate cells. Furthermore, pPB-HSA was able to reduce PDGF-BB-induced fibroblast proliferation in vitro, and proved to be devoid of proliferation-inducing activity itself. We assessed the distribution of pPB-HSA in vivo in two models of fibrosis and related the distribution of pPB-HSA to PDGF-beta receptor density. In rats with liver fibrosis (bile duct ligation model), pPB-HSA quickly accumulated in the liver in contrast to unmodified HSA (P<0.001). The major part of pPB-HSA in the fibrotic liver was localized in hepatic stellate cells. In rats with renal fibrosis (anti-Thy1.1 model), pPB-HSA also homed to the cells that expressed the PDGF-beta receptor, i.e. the mesangial cells in the glomeruli of the kidney. These results indicate that pPB-HSA may be applied as a macromolecular drug-carrier that accumulates specifically in cells expressing the PDGF-beta receptor, thus allowing a selective delivery of anti-fibrotic agents to these cells.


Subject(s)
Fibroblasts/metabolism , Liver Cirrhosis/pathology , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Serum Albumin/metabolism , 3T3 Cells , Animals , Cell Division/drug effects , Fibroblasts/drug effects , Fibrosis/pathology , Humans , Male , Mice , Rats , Rats, Wistar , Serum Albumin/pharmacology
19.
Anat Rec A Discov Mol Cell Evol Biol ; 280(1): 808-20, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15382020

ABSTRACT

After receiving information from afferent nerves, the hypothalamus sends signals to peripheral organs, including the liver, to keep homeostasis. There are two ways for the hypothalamus to signal to the peripheral organs: by stimulating the autonomic nerves and by releasing hormones from the pituitary gland. In order to reveal the involvement of the autonomic nervous system in liver function, we focus in this study on autonomic nerves and neuroendocrine connections between the hypothalamus and the liver. The hypothalamus consists of three major areas: lateral, medial, and periventricular. Each area has some nuclei. There are two important nuclei and one area in the hypothalamus that send out the neural autonomic information to the peripheral organs: the ventromedial hypothalamic nucleus (VMH) in the medial area, the lateral hypothalamic area (LHA), and the periventricular hypothalamic nucleus (PVN) in the periventricular area. VMH sends sympathetic signals to the liver via the celiac ganglia, the LHA sends parasympathetic signals to the liver via the vagal nerve, and the PVN integrates information from other areas of the hypothalamus and sends both autonomic signals to the liver. As for the afferent nerves, there are two pathways: a vagal afferent and a dorsal afferent nerve pathway. Vagal afferent nerves are thought to play a role as sensors in the peripheral organs and to send signals to the brain, including the hypothalamus, via nodosa ganglia of the vagal nerve. On the other hand, dorsal afferent nerves are primary sensory nerves that send signals to the brain via lower thoracic dorsal root ganglia. In the liver, many nerves contain classical neurotransmitters (noradrenaline and acetylcholine) and neuropeptides (substance P, calcitonin gene-related peptide, neuropeptide Y, vasoactive intestinal polypeptide, somatostatin, glucagon, glucagon-like peptide, neurotensin, serotonin, and galanin). Their distribution in the liver is species-dependent. Some of these nerves are thought to be involved in the regulation of hepatic function as well as of hemodynamics. In addition to direct neural connections, the hypothalamus can affect metabolic functions by neuroendocrine connections: the hypothalamus-pancreas axis, the hypothalamus-adrenal axis, and the hypothalamus-pituitary axis. In the hypothalamus-pancreas axis, autonomic nerves release glucagon and insulin, which directly enter the liver and affect liver metabolism. In the hypothalamus-adrenal axis, autonomic nerves release catecholamines such as adrenaline and noradrenaline from the adrenal medulla, which also affects liver metabolism. In the hypothalamus-pituitary axis, release of glucocorticoids and thyroid hormones is stimulated by pituitary hormones. Both groups of hormones modulate hepatic metabolism. Taken together, the hypothalamus controls liver functions by neural and neuroendocrine connections.


Subject(s)
Autonomic Pathways/anatomy & histology , Hypothalamus/anatomy & histology , Liver/innervation , Neural Pathways/anatomy & histology , Neurosecretory Systems/anatomy & histology , Adrenal Glands/physiology , Autonomic Pathways/physiology , Glucose/metabolism , Humans , Hypothalamus/physiology , Liver/anatomy & histology , Neuropeptides/physiology , Neurosecretory Systems/physiology , Pancreas/physiology
20.
Eur J Gastroenterol Hepatol ; 24(12): 1370-80, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22895388

ABSTRACT

AIM: Direct and indirect effects of leptin on hepatic stellate cells (HSCs) have been documented in the literature, whereas little is known about leptin's actions on hepatocytes. Leptin mediates its profibrogenic and proinflammatory effects on HSCs in part through the production of intracellular reactive oxygen species (ROS). In this study, we focus our analysis on leptin-induced ROS production in hepatocytes. METHODS: The expression of leptin receptor isoforms on primary mouse liver cells was examined by real-time quantitative-PCR and western blotting. Cultures were exposed to leptin in combination with inhibitors for reduced nicotinamide adenine dinucleotide phosphate (NADPH) oxidase, MAP kinase/ERK kinase 1 (MEK1) or janus kinase 2 (JAK2). ROS levels were quantified by measuring fluorescence. The effects of leptin on hepatocyte functions and programmed cell death were evaluated by fluorescent or luminescent assays. RESULTS: Leptin induced ROS production in primary hepatocytes by 150-450%, compared with a 20-30% increase in HSCs and liver sinusoidal endothelial cells (LSECs). This ROS production could be inhibited by NADPH oxidase, MEK1 and JAK2 inhibitors. Western blotting indicated that mouse HSCs and LSECs mainly express short leptin receptor isoforms, whereas hepatocytes appeared to express both short and long isoform(s). Leptin-induced ROS production in db/db hepatocytes did not differ from wild-type mice. Finally, leptin had no negative influence on primary hepatocyte functions. CONCLUSION: Leptin induced higher ROS levels in primary hepatocytes than in LSECs and HSCs, depending on NADPH oxidase, MEK1 and JAK2 signalling but not on the long leptin receptor isoform. Furthermore, leptin exposure did not influence primary hepatocyte functionality negatively.


Subject(s)
Hepatocytes/metabolism , Leptin/metabolism , Oxidative Stress , Reactive Oxygen Species/metabolism , Albumins/metabolism , Animals , Apoptosis , Blotting, Western , Cells, Cultured , Cytochrome P-450 CYP1A2/metabolism , Enzyme Inhibitors/pharmacology , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Hepatocytes/drug effects , Hepatocytes/pathology , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/metabolism , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 1/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/metabolism , Oxidative Stress/drug effects , Protein Isoforms , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Receptors, Leptin/genetics , Receptors, Leptin/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Time Factors , Urea/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL