Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Dig Dis Sci ; 62(4): 968-978, 2017 04.
Article in English | MEDLINE | ID: mdl-28194671

ABSTRACT

BACKGROUND AND AIMS: Hyperinsulinemia and insulin resistance are hallmark features of nonalcoholic fatty liver disease and steatohepatitis (NASH). It remains unclear whether and how insulin contributes to the development of fibrosis in NASH. In this study, we explored insulin signaling in the regulation of hepatic stellate cell (HSC) activation and the progression of NASH-fibrosis. METHODS: Phosphorylation of Akt and p70S6K were examined in primary HSC and in a rat model of NASH-fibrosis induced by high-fat and high-cholesterol diet for 24 weeks. HSC activation was analyzed for the changes in cell morphology, intracellular lipid droplets, expression of α-SMA and cell proliferation. The serum markers and histology for NASH-fibrosis were also characterized in animals. RESULTS: Insulin enhanced the expression of smooth muscle actin-α in quiescent but not in activated HSC in culture. Insulin-mediated activation of the PI3K/Akt-p70S6K pathway was involved in the regulation of profibrogenic effects of insulin. Although insulin did not stimulate HSC proliferation directly, the insulin-PI3K/Akt-p70S6K pathway was necessary for serum-enhanced cell proliferation during initial HSC activation. In a rat model of NASH-fibrosis induced by high-fat and high-cholesterol diet, hyperinsulinemia is associated with the activation of p70S6K and enhanced fibrosis. CONCLUSION: The insulin-PI3K/Akt-p70S6K pathway plays an important role in the early activation of HSC. The profibrogenic effect of insulin is dependent on the activation stage of HSC. Dysregulation of the insulin pathway likely correlates with the development of fibrosis in NASH, suggesting a potentially novel antifibrotic target of inhibiting insulin signaling in HSC.


Subject(s)
Hepatic Stellate Cells/metabolism , Liver Cirrhosis/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Animals , Cells, Cultured , Diet, High-Fat/adverse effects , Dose-Response Relationship, Drug , Hepatic Stellate Cells/drug effects , Insulin/pharmacology , Insulin/toxicity , Liver Cirrhosis/chemically induced , Male , Non-alcoholic Fatty Liver Disease/chemically induced , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/physiology
2.
Am J Physiol Gastrointest Liver Physiol ; 302(12): G1397-404, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22517766

ABSTRACT

Hepcidin is a hepatocellular hormone that inhibits the release of iron from certain cell populations, including enterocytes and reticuloendothelial cells. The regulation of hepcidin (HAMP) gene expression by iron status is mediated in part by the signaling molecule bone morphogenetic protein 6 (BMP6). We took advantage of the low iron status of juvenile mice to characterize the regulation of Bmp6 and Hamp1 expression by iron administered in three forms: 1) ferri-transferrin (Fe-Tf), 2) ferric ammonium citrate (FAC), and 3) liver ferritin. Each of these forms of iron enters cells by distinct mechanisms and chemical forms. Iron was parenterally administered to 10-day-old mice, and hepatic expression of Bmp6 and Hamp1 mRNAs was measured 6 h later. We observed that hepatic Bmp6 expression increased in response to ferritin but was unchanged by Fe-Tf or FAC. Hepatic Hamp1 expression likewise increased in response to ferritin and Fe-Tf but was decreased by FAC. Exogenous ferritin increased Bmp6 and Hamp1 expression in older mice as well. Removing iron from ferritin markedly decreased its effect on Bmp6 expression. Exogenously administered ferritin and the derived iron localized in the liver primarily to sinusoidal lining cells. Moreover, expression of Bmp6 mRNA in isolated adult rodent liver cells was much higher in sinusoidal lining cells than hepatocytes (endothelial >> stellate > Kupffer). We conclude that exogenous iron-containing ferritin upregulates hepatic Bmp6 expression, and we speculate that liver ferritin contributes to regulation of Bmp6 and, thus, Hamp1 genes.


Subject(s)
Antimicrobial Cationic Peptides/metabolism , Bone Morphogenetic Protein 6/metabolism , Ferritins/pharmacology , Liver/drug effects , Transferrin/pharmacology , Up-Regulation/drug effects , Animals , Antimicrobial Cationic Peptides/genetics , Bone Morphogenetic Protein 6/genetics , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepcidins , Liver/metabolism , Mice , Signal Transduction/drug effects
3.
Gastroenterology ; 141(5): 1907-14, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21745449

ABSTRACT

BACKGROUND & AIMS: HFE and transferrin receptor 2 (TFR2) are each necessary for the normal relationship between body iron status and liver hepcidin expression. In murine Hfe and Tfr2 knockout models of hereditary hemochromatosis (HH), signal transduction to hepcidin via the bone morphogenetic protein 6 (Bmp6)/Smad1,5,8 pathway is attenuated. We examined the effect of dietary iron on regulation of hepcidin expression via the Bmp6/Smad1,5,8 pathway using mice with targeted disruption of Tfr2, Hfe, or both genes. METHODS: Hepatic iron concentrations and messenger RNA expression of Bmp6 and hepcidin were compared with wild-type mice in each of the HH models on standard or iron-loading diets. Liver phospho-Smad (P-Smad)1,5,8 and Id1 messenger RNA levels were measured as markers of Bmp/Smad signaling. RESULTS: Whereas Bmp6 expression was increased, liver hepcidin and Id1 expression were decreased in each of the HH models compared with wild-type mice. Each of the HH models also showed attenuated P-Smad1,5,8 levels relative to liver iron status. Mice with combined Hfe/Tfr2 disruption were most affected. Dietary iron loading increased hepcidin and Id1 expression in each of the HH models. Compared with wild-type mice, HH mice demonstrated attenuated (Hfe knockout) or no increases in P-Smad1,5,8 levels in response to dietary iron loading. CONCLUSIONS: These observations show that Tfr2 and Hfe are each required for normal signaling of iron status to hepcidin via the Bmp6/Smad1,5,8 pathway. Mice with combined loss of Hfe and Tfr2 up-regulate hepcidin in response to dietary iron loading without increases in liver Bmp6 messenger RNA or steady-state P-Smad1,5,8 levels.


Subject(s)
Antimicrobial Cationic Peptides/metabolism , Iron, Dietary/pharmacology , Membrane Proteins/deficiency , Receptors, Transferrin/deficiency , Signal Transduction/physiology , Smad1 Protein/metabolism , Smad5 Protein/metabolism , Smad8 Protein/metabolism , Animals , Bone Morphogenetic Protein 6/metabolism , Hemochromatosis Protein , Hepcidins , Histocompatibility Antigens Class I/genetics , Inhibitor of Differentiation Protein 1/metabolism , Iron/metabolism , Liver/drug effects , Liver/metabolism , Membrane Proteins/genetics , Mice , Mice, Knockout , Models, Animal , RNA, Messenger/metabolism , Receptors, Transferrin/genetics , Up-Regulation/drug effects , Up-Regulation/physiology
4.
Annu Rev Nutr ; 31: 117-37, 2011 Aug 21.
Article in English | MEDLINE | ID: mdl-21548776

ABSTRACT

Murine models have made valuable contributions to our understanding of iron metabolism. Investigation of mice with inherited forms of anemia has led to the discovery of novel proteins involved in iron homeostasis. A growing number of murine models are being developed to investigate mitochondrial iron metabolism. Mouse strains are available for the major forms of hereditary hemochromatosis. Findings in murine models support the concept that the pathogenesis of nearly all forms of hereditary hemochromatosis involves inappropriately low expression of hepcidin. The availability of mice with floxed iron-related genes allows the study of the in vivo consequences of cell-selective deletion of these genes.


Subject(s)
Disease Models, Animal , Homeostasis , Iron/metabolism , Mice , Models, Animal , Anemia/genetics , Anemia/metabolism , Animals , Hemochromatosis/genetics , Hemochromatosis/metabolism , Mice, Knockout , Mitochondria/metabolism , Signal Transduction
5.
J Hepatol ; 52(3): 425-31, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20133002

ABSTRACT

BACKGROUND & AIMS: Hereditary haemochromatosis type 3 is caused by mutations in transferrin receptor (TFR) 2. TFR2 has been shown to mediate iron transport in vitro and regulate iron homeostasis. The aim of this study was to determine the role of Tfr2 in iron transport in vivo using a Tfr2 mutant mouse. METHODS: Tfr2 mutant and wild-type mice were injected intravenously with (59)Fe-transferrin and tissue (59)Fe uptake was measured. Tfr1, Tfr2 and ferroportin expression was measured by real-time PCR and Western blot. Cellular localisation of ferroportin was determined by immunohistochemistry. RESULTS: Transferrin-bound iron uptake by the liver and spleen in Tfr2 mutant mice was reduced by 20% and 65%, respectively, whilst duodenal and renal uptake was unchanged compared with iron-loaded wild-type mice. In Tfr2 mutant mice, liver Tfr2 protein was absent, whilst ferroportin protein was increased in non-parenchymal cells and there was a low level of expression in hepatocytes. Tfr1 expression was unchanged compared with iron-loaded wild-type mice. Splenic Tfr2 protein expression was absent whilst Tfr1 and ferroportin protein expression was increased in Tfr2 mutant mice compared with iron-loaded wild-type mice. CONCLUSIONS: A small reduction in hepatic transferrin-bound iron uptake in Tfr2 mutant mice suggests that Tfr2 plays a minor role in liver iron transport and its primary role is to regulate iron metabolism. Increased ferroportin expression due to decreased hepcidin mRNA levels is likely to be responsible for impaired splenic iron uptake in Tfr2 mutant mice.


Subject(s)
Hemochromatosis/metabolism , Iron/metabolism , Receptors, Transferrin/metabolism , Transferrin/metabolism , Animals , Antimicrobial Cationic Peptides/metabolism , Biological Transport/physiology , Cation Transport Proteins/metabolism , Disease Models, Animal , Female , Hemochromatosis/genetics , Hepcidins , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , RNA, Messenger/metabolism , Receptors, Transferrin/genetics , Spleen/metabolism
6.
PLoS One ; 4(9): e7212, 2009 Sep 29.
Article in English | MEDLINE | ID: mdl-19787063

ABSTRACT

Iron is an essential trace element whose absorption is usually tightly regulated in the duodenum. HFE-related hereditary hemochromatosis (HH) is characterized by abnormally low expression of the iron-regulatory hormone, hepcidin, which results in increased iron absorption. The liver is crucial for iron homeostasis as it is the main production site of hepcidin. The aim of this study was to explore and compare the genome-wide transcriptome response to Hfe deficiency and dietary iron overload in murine liver and duodenum. Illumina arrays containing over 47,000 probes were used to study global transcriptional changes. Quantitative RT-PCR (Q-RT-PCR) was used to validate the microarray results. In the liver, the expression of 151 genes was altered in Hfe(-/-) mice while dietary iron overload changed the expression of 218 genes. There were 173 and 108 differentially expressed genes in the duodenum of Hfe(-/-) mice and mice with dietary iron overload, respectively. There was 93.5% concordance between the results obtained by microarray analysis and Q-RT-PCR. Overexpression of genes for acute phase reactants in the liver and a strong induction of digestive enzyme genes in the duodenum were characteristic of the Hfe-deficient genotype. In contrast, dietary iron overload caused a more pronounced change of gene expression responsive to oxidative stress. In conclusion, Hfe deficiency caused a previously unrecognized increase in gene expression of hepatic acute phase proteins and duodenal digestive enzymes.


Subject(s)
Animal Feed , Duodenum/metabolism , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , Iron Overload/genetics , Liver/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Transcription, Genetic , Animals , Antimicrobial Cationic Peptides/metabolism , Genome-Wide Association Study , Hemochromatosis Protein , Hepcidins , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Oxidative Stress
7.
BMC Neurosci ; 10: 36, 2009 Apr 22.
Article in English | MEDLINE | ID: mdl-19386095

ABSTRACT

BACKGROUND: Defective iron homeostasis may be involved in the development of some diseases within the central nervous system. Although the expression of genes involved in normal iron balance has been intensively studied in other tissues, little is known about their expression in the brain. We investigated the mRNA levels of hepcidin (HAMP), HFE, neogenin (NEO1), transferrin receptor 1 (TFRC), transferrin receptor 2 (TFR2), and hemojuvelin (HFE2) in normal human brain, brain tumors, and astrocytoma cell lines. The specimens included 5 normal brain tissue samples, 4 meningiomas, one medulloblastoma, 3 oligodendrocytic gliomas, 2 oligoastrocytic gliomas, 8 astrocytic gliomas, and 3 astrocytoma cell lines. RESULTS: Except for hemojuvelin, all genes studied had detectable levels of mRNA. In most tumor types, the pattern of gene expression was diverse. Notable findings include high expression of transferrin receptor 1 in the hippocampus and medulla oblongata compared to other brain regions, low expression of HFE in normal brain with elevated HFE expression in meningiomas, and absence of hepcidin mRNA in astrocytoma cell lines despite expression in normal brain and tumor specimens. CONCLUSION: These results indicate that several iron-related genes are expressed in normal brain, and that their expression may be dysregulated in brain tumors.


Subject(s)
Brain Neoplasms/metabolism , Brain/metabolism , Gene Expression Regulation, Neoplastic/physiology , Histocompatibility Antigens Class I/metabolism , Membrane Proteins/metabolism , Adult , Aged , Aged, 80 and over , Antigens, CD/genetics , Antigens, CD/metabolism , Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/metabolism , Astrocytoma/genetics , Astrocytoma/metabolism , Brain Neoplasms/genetics , Cell Line, Tumor , Female , GPI-Linked Proteins , Hemochromatosis Protein , Hepcidins , Histocompatibility Antigens Class I/genetics , Humans , Male , Membrane Proteins/genetics , Meningioma/genetics , Meningioma/metabolism , Middle Aged , Oligodendroglioma/genetics , Oligodendroglioma/metabolism , RNA, Messenger/analysis , Receptors, Transferrin/genetics , Receptors, Transferrin/metabolism , Statistics, Nonparametric , Young Adult
8.
Hepatology ; 48(3): 991-1001, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18752323

ABSTRACT

Following the discovery of the HFE gene in 1996 and its linkage to the iron overload disorder hereditary hemochromatosis (HH) there have been profound developments in our understanding of the pathogenesis of the biochemical and clinical manifestations of a number of iron overload disorders. This article provides an update of recent developments and key issues relating to iron homeostasis and inherited disorders of iron overload, with emphasis on HFE-related HH, and is based on the content of the American Association for the Study of Liver Diseases Single-Topic Conference entitled "Hemochromatosis: What has Happened After HFE?" which was held at the Emory Convention Center in Atlanta, September 7-9, 2007.


Subject(s)
Hemochromatosis/genetics , Histocompatibility Antigens Class I/genetics , Membrane Proteins/genetics , Genetic Testing , Hemochromatosis/diagnosis , Hemochromatosis/metabolism , Hemochromatosis Protein , Histocompatibility Antigens Class I/metabolism , Homeostasis , Humans , Iron/metabolism , Liver/metabolism , Liver/pathology , Membrane Proteins/metabolism
9.
Arch Biochem Biophys ; 474(1): 193-7, 2008 Jun 01.
Article in English | MEDLINE | ID: mdl-18353247

ABSTRACT

Mutations in either HFE or transferrin receptor 2 (TfR2) cause decreased expression of the iron regulatory hormone hepcidin and hemochromatosis. HFE and TfR2 were recently discovered to form a stable complex at the cell membrane when co-expressed in heterologous cell lines. We analyzed the functional consequences of the co-expression of these proteins using transfected TRVb cells, a Chinese hamster ovary derived cell line without endogenous HFE or transferrin receptor. The co-expression of TfR2 in TRVb cells expressing HFE led to accelerated HFE biosynthesis and late-Golgi maturation, suggesting interaction prior to cell surface localization. The co-expression of HFE in cells expressing TfR2 led to increased affinity for diferric transferrin, increased transferrin-dependent iron uptake, and relative resistance to iron chelation. These observations indicate that HFE influences the functional properties of TfR2, and suggests a model in which the interaction of these proteins might influence signal transduction to hepcidin.


Subject(s)
Endocytosis , Histocompatibility Antigens Class I/metabolism , Iron/metabolism , Membrane Proteins/metabolism , Receptors, Transferrin/metabolism , Transferrin/metabolism , Blotting, Western , Cell Line , Cell Membrane/metabolism , Electrophoresis, Polyacrylamide Gel , Golgi Apparatus/metabolism , Hemochromatosis Protein , Humans , Immunoprecipitation
11.
BMC Genomics ; 8: 379, 2007 Oct 19.
Article in English | MEDLINE | ID: mdl-17949489

ABSTRACT

BACKGROUND: Hereditary hemochromatosis (HH) encompasses genetic disorders of iron overload characterized by deficient expression or function of the iron-regulatory hormone hepcidin. Mutations in 5 genes have been linked to this disease: HFE, TFR2 (encoding transferrin receptor 2), HAMP (encoding hepcidin), SLC40A1 (encoding ferroportin) and HJV (encoding hemojuvelin). Hepcidin inhibits iron export from cells into plasma. Hemojuvelin, an upstream regulator of hepcidin expression, is expressed in mice mainly in the heart and skeletal muscle. It has been suggested that soluble hemojuvelin shed by the muscle might reach the liver to influence hepcidin expression. Heart muscle is one of the target tissues affected by iron overload, with resultant cardiomyopathy in some HH patients. Therefore, we investigated the effect of iron overload on gene expression in skeletal muscle and heart using Illuminatrade mark arrays containing over 47,000 probes. The most apparent changes in gene expression were confirmed using real-time RT-PCR. RESULTS: Genes with up-regulated expression after iron overload in both skeletal and heart muscle included angiopoietin-like 4, pyruvate dehydrogenase kinase 4 and calgranulin A and B. The expression of transferrin receptor, heat shock protein 1B and DnaJ homolog B1 were down-regulated by iron in both muscle types. Two potential hepcidin regulatory genes, hemojuvelin and neogenin, showed no clear change in expression after iron overload. CONCLUSION: Microarray analysis revealed iron-induced changes in the expression of several genes involved in the regulation of glucose and lipid metabolism, transcription and cellular stress responses. These may represent novel connections between iron overload and pathological manifestations of HH such as cardiomyopathy and diabetes.

12.
Liver Int ; 27(8): 1066-75, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17845534

ABSTRACT

BACKGROUND/AIMS: Hepatic stellate cells (HSC) play a key role in hepatic fibrogenesis and thus, it is important to understand the intracellular signalling pathways that influence their behaviour. This study investigated the expression and regulation of protein kinase C (PKC) in HSC. RESULTS: Western blot analysis indicates that rat HSC express at least four PKC isoforms, PKC-alpha, PKC-delta, PKC-epsilon and PKC-zeta. PKC-alpha and PKC-zeta were located predominantly in the cytosol and were redistributed to the membrane by the PKC agonist, phorbol 12-myristate 13-acetate (PMA), while PKC-delta and PKC-epsilon were highly membrane-bound and did not undergo translocation by PMA. PKC-alpha, PKC-delta and PKC-zeta were rapidly downregulated by PMA. However, PKC-epsilon was resistant to downregulation. We also examined phosphorylation of myristoylated alanine-rich C kinase substrate (MARCKS), a specific substrate of PKC, as another approach to assess activation of PKC. Platelet-derived growth factor (PDGF) and PMA increased the phosphorylation of MARCKS, suggesting that PDGF can induce PKC activation. PDGF-induced stimulation of extracellular signal-regulated kinase, phosphatidylinositol 3-kinase and p70-S6 kinase was not abrogated by downregulation of PKC-alpha, PKC-delta and PKC-zeta. Prolonged PKC inhibition did not inhibit the fibrogenic phenotype. CONCLUSION: Multiple PKC isoforms are expressed in rat HSC and are differentially regulated by PMA. PDGF activates certain mitogenic signalling pathways independent of PKC-alpha, PKC-delta and PKC-zeta. Specific PKC isoforms may modulate different cell functions in HSC.


Subject(s)
Liver/drug effects , Platelet-Derived Growth Factor/metabolism , Protein Kinase C/metabolism , Signal Transduction/drug effects , Tetradecanoylphorbol Acetate/pharmacology , Actins/metabolism , Animals , Becaplermin , Carbazoles/pharmacology , Cell Differentiation/drug effects , Cell Membrane/drug effects , Cell Membrane/enzymology , Cells, Cultured , Collagen Type I/metabolism , Cytosol/drug effects , Cytosol/enzymology , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Indoles/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism , Isoenzymes/metabolism , Liver/cytology , Liver/enzymology , Liver/metabolism , Male , Maleimides/pharmacology , Membrane Proteins/metabolism , Molecular Chaperones/metabolism , Myristoylated Alanine-Rich C Kinase Substrate , Phenotype , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Protein Kinase C/antagonists & inhibitors , Protein Kinase C-alpha/metabolism , Protein Kinase C-delta/metabolism , Protein Kinase C-epsilon/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Transport , Proto-Oncogene Proteins c-sis , Rats , Rats, Sprague-Dawley , Ribosomal Protein S6 Kinases, 70-kDa/metabolism
13.
Liver Int ; 26(5): 559-65, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16762000

ABSTRACT

BACKGROUND AND AIMS: Endotoxin-responsive monocytes/macrophages (CD14-positive) are potential sources of profibrogenic factors. The aims of this study were to determine (1) whether hepatic CD14-positive cells are present in various forms of chronic liver disease, and (2) the relationship between CD14-positive cells, myofibroblasts, and fibrosis in these diseases. METHODS: Liver specimens from control subjects (n = 12) and those with primary biliary cirrhosis (n = 18), chronic hepatitis C (n = 13), or nonalcoholic steatohepatitis (n = 13) were immunostained for CD14, CD68, and alpha-smooth muscle actin (SMA) and the number of cells expressing these antigens was determined. Fibrosis and inflammation were also assessed. RESULTS: The total number of hepatic CD68-positive cells was similar in diseased and control livers. The number of CD14-positive cells was increased in advanced fibrosis in primary biliary cirrhosis and hepatitis C but not in nonalcoholic steatohepatitis. The number of CD14-positive cells was also increased in hepatitis C specimens with high inflammatory activity. CD14-positive cells were often associated with alpha-SMA-positive myofibroblasts in fibrous septa. CONCLUSIONS: The number of hepatic CD14-positive cells is increased in advanced fibrosis in subjects with primary biliary cirrhosis and hepatitis C but not in nonalcoholic steatohepatitis. In primary biliary cirrhosis and hepatitis C, CD14-positive macrophages are found in close proximity to fibrous septa and myofibroblasts. In hepatitis C, an increased number of CD14-positive cells are associated with high inflammatory activity.


Subject(s)
Hepatitis/pathology , Lipopolysaccharide Receptors , Liver Diseases/pathology , Macrophages/immunology , Biopsy , Case-Control Studies , Chronic Disease , Fatty Liver/immunology , Fatty Liver/pathology , Female , Hepatitis/immunology , Hepatitis C, Chronic/immunology , Hepatitis C, Chronic/pathology , Humans , Inflammation , Liver/immunology , Liver/pathology , Liver Cirrhosis, Biliary/immunology , Liver Cirrhosis, Biliary/pathology , Liver Diseases/immunology , Male , Middle Aged , Monocytes/immunology
14.
J Lab Clin Med ; 147(5): 234-41, 2006 May.
Article in English | MEDLINE | ID: mdl-16697771

ABSTRACT

Proliferation of activated hepatic stellate cells (HSC) is an important event in the development of hepatic fibrosis. Insulin-like growth factor-1 (IGF-1) has been shown to be mitogenic for HSC, but the intracellular signaling pathways involved have not been fully characterized. Thus, the aims of the current study were to examine the roles of the extracellular signal-regulated kinase (ERK), phosphatidylinositol 3-kinase (PI3-K) and p70-S6 kinase (p70-S6-K) signaling pathways in IGF-1- and platelet-derived growth factor (PDGF)-induced mitogenic signaling of HSC and to examine the potential crosstalk between these pathways. Both IGF-1 and PDGF increased ERK, PI3-K and p70-S6-K activity. When evaluating potential crosstalk between these signaling pathways, we observed that PI3-K is required for p70-S6-K activation by IGF-1 and PDGF, and is partially responsible for PDGF-induced ERK activation. PDGF and IGF-1 also increased the levels of cyclin D1 and phospho-glycogen synthase kinase-3beta. Coordinate activation of ERK, PI3-K and p70-S6-K is important for perpetuating the activated state of HSC during fibrogenesis.


Subject(s)
Insulin-Like Growth Factor I/pharmacology , Kupffer Cells/drug effects , Platelet-Derived Growth Factor/pharmacology , Signal Transduction/drug effects , Animals , Cell Proliferation/drug effects , Cells, Cultured , Cyclin D1/antagonists & inhibitors , Cyclin D1/biosynthesis , Drug Combinations , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/biosynthesis , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/biosynthesis , Glycogen Synthase Kinase 3 beta , Kupffer Cells/enzymology , Male , Phosphatidylinositol 3-Kinases/biosynthesis , Phosphoinositide-3 Kinase Inhibitors , Rats , Rats, Sprague-Dawley , Ribosomal Protein S6 Kinases, 70-kDa/antagonists & inhibitors , Ribosomal Protein S6 Kinases, 70-kDa/biosynthesis
15.
Am J Physiol Gastrointest Liver Physiol ; 290(4): G590-4, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16537971

ABSTRACT

The majority of clinical cases of iron overload is caused by mutations in the HFE gene. However, the role that HFE plays in the physiology of intestinal iron absorption remains enigmatic. Two major models have been proposed: 1) HFE exerts its effects on iron homeostasis indirectly, by modulating the expression of hepcidin; and 2) HFE exerts its effects directly, by changing the iron status (and therefore the iron absorptive activity) of intestinal enterocytes. The first model places the primary role of HFE in the liver (hepatocytes and/or Kupffer cells). The second model places the primary role in the duodenum (crypt cells or villus enterocytes). These models are not mutually exclusive, and it is possible that HFE influences the iron status in each of these cell populations, leading to cell type-specific downstream effects on intestinal iron absorption and body iron distribution.


Subject(s)
Antimicrobial Cationic Peptides/metabolism , Histocompatibility Antigens Class I/metabolism , Intestinal Absorption/physiology , Iron/metabolism , Liver/metabolism , Membrane Proteins/metabolism , Models, Biological , Animals , Hemochromatosis Protein , Hepcidins , Humans
16.
Semin Liver Dis ; 25(4): 411-9, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16315135

ABSTRACT

Hereditary hemochromatosis (HH) encompasses several inherited disorders of iron homeostasis characterized by increased gastrointestinal iron absorption and tissue iron deposition. The most common form of this disorder is HFE-related HH, nearly always caused by homozygosity for the C282Y mutation. A substantial proportion of C282Y homozygotes do not develop clinically significant iron overload, suggesting roles for environmental factors and modifier genes in determining the phenotype. Recent studies have demonstrated that the pathogenesis of nearly all forms of HH involves inappropriately decreased expression of the iron-regulatory hormone hepcidin. Hepcidin serves to decrease the export of iron from reticuloendothelial cells and absorptive enterocytes. Thus, HH patients demonstrate increased iron release from these cell types, elevated circulating iron, and iron deposition in vulnerable tissues. The mechanism by which HFE influences hepcidin expression is an area of current investigation and may offer insights into the phenotypic variability observed in persons with mutations in HFE.


Subject(s)
Hemochromatosis/physiopathology , Absorption , Antimicrobial Cationic Peptides/biosynthesis , Antimicrobial Cationic Peptides/genetics , Gene Expression/physiology , Hemochromatosis/genetics , Hemochromatosis Protein , Hepcidins , Histocompatibility Antigens Class I/biosynthesis , Histocompatibility Antigens Class I/genetics , Humans , Intestinal Mucosa/metabolism , Iron/metabolism , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Mutation
17.
Am J Gastroenterol ; 100(4): 837-41, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15784029

ABSTRACT

OBJECTIVES: Hepatic fibrosis is a complication of hereditary hemochromatosis. The aim of this study was to determine whether the product of the magnitude and duration of hepatic iron exposure is related to the risk of significant fibrosis. METHODS: Receiver-operating characteristic curve analysis to determine the utility of hepatic iron concentration (HIC) and age in the diagnosis of low- or high-grade fibrosis was undertaken retrospectively in 60 subjects who had undergone liver biopsy for assessment of hereditary hemochromatosis. A prospective pilot study was then conducted in 10 additional subjects to evaluate utility of magnetic resonance imaging (MRI) measurements of HIC to predict fibrosis. RESULTS: Eighteen subjects had high-grade fibrosis while 42 subjects had low-grade fibrosis. Hepatic iron concentration alone was highly sensitive (100%) but of limited specificity (67%) in diagnosis of high-grade fibrosis. The product of [HIC x age] had a sensitivity and specificity of 100% and 86%, respectively, for diagnosis of high-grade fibrosis. Magnetic resonance imaging measurements also provided accurate assignment of subjects into fibrosis severity groups. CONCLUSIONS: Duration of exposure to iron is important in the development of hepatic fibrosis in hereditary hemochromatosis. The product of HIC and age is highly sensitive and specific for diagnosis of high-grade fibrosis and can be obtained using MRI.


Subject(s)
Hemochromatosis/genetics , Image Processing, Computer-Assisted , Iron/metabolism , Liver Cirrhosis/genetics , Liver/pathology , Magnetic Resonance Imaging , Adult , Age Factors , Biopsy , DNA Mutational Analysis , Disease Progression , Female , Ferritins/blood , Hemochromatosis/complications , Hemochromatosis/diagnosis , Hemochromatosis/pathology , Hemochromatosis Protein , Histocompatibility Antigens Class I/genetics , Homozygote , Humans , Liver Cirrhosis/classification , Liver Cirrhosis/diagnosis , Liver Cirrhosis/pathology , Male , Membrane Proteins/genetics , Middle Aged , Pilot Projects , Prospective Studies , Retrospective Studies , Risk , Sensitivity and Specificity
19.
Liver Int ; 24(5): 446-51, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15482341

ABSTRACT

BACKGROUND/AIMS: Iron overload in hereditary hemochromatosis (HH) may result in hepatic fibrosis and cirrhosis, primarily due to collagen production by hepatic stellate cells that become activated to myofibroblasts. Endotoxin-responsive monocytes/macrophages (CD14-positive) are potential sources of profibrogenic factors. The aims of this study were to determine (1) whether CD14-positive monocytes/macrophages are present in the livers of patients with HH and (2) the potential relationship between CD14-positive cells and hepatic fibrosis in HH. METHODS: HH was diagnosed using standard clinical, biochemical and genotypic parameters. Liver specimens from HH patients and control subjects were immunostained for CD14, CD68 and alpha-smooth muscle actin (alpha-SMA) and the number of cells expressing these antigens was determined. Fibrosis was assessed by routine histological methods. RESULTS: The total number of hepatic CD68-positive monocytes/macrophages was similar in HH patients and control subjects; however, there was a nine-fold increase in the number of CD14-positive monocytes/macrophages in HH patients. Control subjects had very low levels of hepatic CD14 expression. In HH livers with advanced fibrosis, CD14-positive monocytes/macrophages were often associated with fibrous septa containing myofibroblasts expressing alpha-SMA. CONCLUSIONS: There was a substantial increase in hepatic CD14-positive monocytes/macrophages in HH and, in livers with advanced fibrosis, these cells were often associated with fibrous septa and septal myofibroblasts. The total number of monocytes/macrophages was similar in HH and control livers. In control human liver, Kupffer cells had a very low expression of CD14. These findings suggest that CD14-positive monocytes/macrophages may contribute to the process of hepatic fibrogenesis in HH.


Subject(s)
Hemochromatosis , Liver Cirrhosis/pathology , Liver/pathology , Macrophages/pathology , Monocytes/pathology , Adult , Aged , Biomarkers/analysis , Cell Count , Hemochromatosis/complications , Hemochromatosis/genetics , Hemochromatosis/pathology , Humans , Immunoenzyme Techniques , Kupffer Cells/immunology , Kupffer Cells/pathology , Lipopolysaccharide Receptors/metabolism , Liver/immunology , Liver Cirrhosis/etiology , Macrophages/immunology , Male , Middle Aged , Monocytes/immunology
20.
Clin Liver Dis ; 8(4): 755-73, vii, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15464654

ABSTRACT

Hereditary hemochromatosis comprises several inherited disorders of iron homeostasis characterized by increased gastrointestinal iron absorpstion and resultant tissue iron deposition. The identification of HFE and other genes involved in iron metabolism has greatly expanded our understanding of hereditary hemochromatosis. Two major hypotheses have been proposed to explain the pathogenesis of HFE-related hereditary hemochromatosis: the hepcidin hypothesis and the duodenal crypt cell programming hypothesis.


Subject(s)
Genetic Predisposition to Disease , Hemochromatosis/genetics , Hemochromatosis/physiopathology , Iron/metabolism , Iron/pharmacokinetics , Animals , Antimicrobial Cationic Peptides/pharmacology , Disease Models, Animal , Duodenum/cytology , Duodenum/physiology , Hepcidins , Humans , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...