Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Transplant ; 29: 963689720950221, 2020.
Article in English | MEDLINE | ID: mdl-32813573

ABSTRACT

Human amnion epithelial cells (hAECs) exert potent antifibrotic and anti-inflammatory effects when transplanted into preclinical models of tissue fibrosis. These effects are mediated in part via the secretion of soluble factors by hAECs which modulate signaling pathways and affect cell types involved in inflammation and fibrosis. Based on these reports, we hypothesized that these soluble factors may also support liver regeneration during chronic liver injury. To test this, we characterized the effect of both hAECs and hAEC-conditioned medium (CM) on liver repair in a mouse model of carbon tetrachloride (CCl4)-induced fibrosis. Liver repair was assessed by liver fibrosis, hepatocyte proliferation, and the liver progenitor cell (LPC) response. We found that the administration of hAECs or hAEC-CM reduced liver injury and fibrosis, sustained hepatocyte proliferation, and reduced LPC numbers during chronic liver injury. Additionally, we undertook in vitro studies to document both the cell-cell and paracrine-mediated effects of hAECs on LPCs by investigating the effects of co-culturing the LPCs and hAECs and hAEC-CM on LPCs. We found little change in LPCs co-cultured with hAECs. In contrast, hAEC-CM enhances LPC proliferation and differentiation. These findings suggest that paracrine factors secreted by hAECs enhance liver repair by reducing fibrosis while promoting regeneration during chronic liver injury.


Subject(s)
Amnion/metabolism , Epithelial Cells/metabolism , Liver Cirrhosis/physiopathology , Liver Cirrhosis/therapy , Liver Regeneration , Liver/injuries , Animals , Cell Count , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Culture Media, Conditioned/pharmacology , Disease Models, Animal , Epithelial Cells/drug effects , Gene Expression Regulation/drug effects , Gene Ontology , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Inflammation/pathology , Liver/drug effects , Liver/pathology , Liver Regeneration/drug effects , Macrophages/drug effects , Macrophages/pathology , Male , Metabolic Networks and Pathways/drug effects , Mice, Inbred C57BL , Solubility , Stem Cells/drug effects , Stem Cells/metabolism , Stem Cells/pathology
2.
World J Gastroenterol ; 23(31): 5692-5699, 2017 Aug 21.
Article in English | MEDLINE | ID: mdl-28883694

ABSTRACT

AIM: To evaluate the role of tissue factor (TF) and protease activated receptor (PAR)-2 in liver fibrosis. METHODS: Using CCl4 administration for eight weeks, we induced hepatic fibrosis in wild-type C57BL/6 mice and in mice with deletion of the cytoplasmic signalling domain of TF (TF§CT/§CT), deletion of PAR-2 (PAR-2-/-) and combined deletion of TF signalling domain and PAR-2 (TF§CT/§CT/PAR-2-/-). Hepatic fibrosis area was assessed by quantitative imaging of picrosirius red staining. Hepatic collagen content was assessed by hydroxyproline levels. Hepatic stellate cells (αSMA positive) and hepatic macrophages (CD68 positive) were identified by immunohistochemistry. Hepatic gene expression was determined by PCR and liver TGFß1 content by ELISA. RESULTS: CCl4 treated mice with deletion of the PAR-2 gene (PAR-2-/-) and the cytoplasmic domain of TF (TF§CT/§CT) developed significantly less hepatic fibrosis, characterised by reduced liver fibrosis area and hydroxyproline content, compared to control wildtype mice treated with CCl4. The observed reduction in histological fibrosis was accompanied by a significant decrease in the hepatic content of TGFß, the prototypic fibrogenic cytokine, as well as fewer activated hepatic stellate cells and hepatic macrophages. Deletion of the TF cytoplasmic signalling domain reduced hepatic fibrosis to levels similar to those observed in mice lacking PAR-2 signalling but combined deletion provided no added protection against fibrosis indicating a lack of mutual modulating effects that have been observed in other contexts such as angiogenic responses. CONCLUSION: Tissue factor cytoplasmic domain is involved in TF-PAR-2 signalling initiating hepatic fibrosis and is a potential therapeutic target, as its deletion would not impact coagulation.


Subject(s)
Liver Cirrhosis, Experimental/pathology , Liver/pathology , Protein Domains/genetics , Receptor, PAR-2/metabolism , Thromboplastin/metabolism , Amino Acid Sequence/genetics , Animals , Blood Coagulation , Carbon Tetrachloride/toxicity , Collagen/analysis , Hepatic Stellate Cells/immunology , Humans , Hydroxyproline/analysis , Liver/cytology , Liver/immunology , Liver Cirrhosis, Experimental/blood , Liver Cirrhosis, Experimental/chemically induced , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, PAR-2/genetics , Sequence Deletion , Signal Transduction , Thromboplastin/genetics , Transforming Growth Factor beta/analysis
3.
Cytotherapy ; 16(8): 1132-44, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24642017

ABSTRACT

BACKGROUND: Intravenous infusion of human amniotic epithelial cells (hAECs) has been shown to ameliorate hepatic fibrosis in murine models. Hepatic stellate cells (HSCs) are the principal collagen-secreting cells in the liver. The aim of this study was to investigate whether factors secreted by hAECs and present in hAEC-conditioned medium (CM) have anti-fibrotic effects on activated human HSCs. METHODS: Human AECs were isolated from the placenta and cultured. Human hepatic stellate cells were exposed to hAEC CM to determine potential anti-fibrotic effects. RESULTS: HSCs treated for 48 h with hAEC CM displayed a significant reduction in the expression of the myofibroblast markers α-smooth muscle actin and platelet-derived growth factor. Expression of the pro-fibrotic cytokine transforming growth factor-ß1 (TGF-ß1) and intracellular collagen were reduced by 45% and 46%, respectively. Human AEC CM induced HSC apoptosis in 11.8% of treated cells and reduced HSC proliferation. Soluble human leukocyte antigen-G1, a hAEC-derived factor, significantly decreased TGF-ß1 and collagen production in activated HSCs, although the effect on collagen production was less than that of hAEC CM. The reduction in collagen and TGF-B1 could not be attributed to PGE2, relaxin, IL-10, TGF-B3, FasL or TRAIL. CONCLUSIONS: Human AEC CM treatment suppresses markers of activation, proliferation and fibrosis in human HSCs as well as inducing apoptosis and reducing proliferation. Human AEC CM treatment may be effective in ameliorating liver fibrosis and warrants further study.


Subject(s)
Amniotic Fluid/cytology , Collagen/biosynthesis , Culture Media, Conditioned/pharmacology , Hepatic Stellate Cells/drug effects , Amniotic Fluid/metabolism , Animals , Apoptosis/drug effects , Collagen/metabolism , Disease Models, Animal , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gene Expression Regulation/drug effects , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Humans , Transforming Growth Factor beta/biosynthesis
4.
PLoS One ; 7(6): e38631, 2012.
Article in English | MEDLINE | ID: mdl-22719909

ABSTRACT

Chronic hepatic inflammation from multiple etiologies leads to a fibrogenic response that can progress to cirrhosis and liver failure. Transplantation of human amniotic epithelial cells (hAEC) from term delivered placenta has been shown to decrease mild to moderate hepatic fibrosis in a murine model. To model advanced human liver disease and assess the efficacy of hAEC therapy, we transplanted hAEC in mice with advanced hepatic fibrosis. Immunocompetent C57BL/6 mice were administered carbon tetrachloride (CCl(4)) twice weekly resulting in bridging fibrosis by 12 weeks. hAEC (2 × 10(6)) were infused via the tail vein at week 8 or weeks 8 and 10 (single and double dose, respectively). Human cells were detected in mouse liver four weeks after transplantation showing hAEC engraftment. CCl(4) treated mice receiving single or double hAEC doses showed a significant but similar decrease in liver fibrosis area associated with decreased activation of collagen-producing hepatic stellate cells and decreased hepatic protein levels of the pro-fibrogenic cytokine, transforming growth factor-beta1. CCl(4) administration caused hepatic T cell infiltration that decreased significantly following hAEC transplantation. Hepatic macrophages play a crucial role in both fibrogenesis and fibrosis resolution. Mice exposed to CCl(4) demonstrated increased numbers of hepatic macrophages compared to normal mice; the number of macrophages decreased significantly in CCl(4) treated mice given hAEC. These mice had significantly lower hepatic protein levels of the chemokine monocyte chemoattractant protein-1 than mice given CCl(4) alone. Alternatively activated M2 macrophages are associated with fibrosis resolution. CCl(4) treated mice given hAEC showed increased expression of genes associated with M2 macrophages including YM-1, IL-10 and CD206. We provide novel data showing that hAEC transplantation induces a wound healing M2 macrophage phenotype associated with reduction of established hepatic fibrosis that justifies further investigation of this potential cell-based therapy for advanced hepatic fibrosis.


Subject(s)
Amnion/transplantation , Cell Transplantation , Liver Cirrhosis/prevention & control , Macrophage Activation , Amnion/cytology , Animals , Antibody Formation , Carbon Tetrachloride/administration & dosage , Carbon Tetrachloride/toxicity , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Epithelial Cells/transplantation , Humans , Immunohistochemistry , Liver Cirrhosis/chemically induced , Liver Cirrhosis/immunology , Male , Mice , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction
5.
Hepatology ; 55(3): 879-87, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22095855

ABSTRACT

UNLABELLED: Protease-activated receptor (PAR) 2 is a G-protein-coupled receptor that is activated after proteolytic cleavage by serine proteases, including mast cell tryptase and activated coagulation factors. PAR-2 activation augments inflammatory and profibrotic pathways through the induction of genes encoding proinflammatory cytokines and extracellular matrix proteins. Thus, PAR-2 represents an important interface linking coagulation and inflammation. PAR-2 is widely expressed in cells of the gastrointestinal tract, including hepatic stellate cells (HSCs), endothelial cells, and hepatic macrophages; however, its role in liver fibrosis has not been previously examined. We studied the development of CCl(4) -induced liver fibrosis in PAR-2 knockout mice, and showed that PAR-2 deficiency reduced the progression of liver fibrosis, hepatic collagen gene expression, and hydroxyproline content. Reduced fibrosis was associated with decreased transforming growth factor beta (TGFß) gene and protein expression and decreased matrix metalloproteinase 2 and tissue inhibitor of matrix metalloproteinase 1 gene expression. In addition, PAR-2 stimulated activation, proliferation, collagen production, and TGFß protein production by human stellate cells, indicating that hepatic PAR-2 activation increases profibrogenic cytokines and collagen production both in vivo and in vitro. CONCLUSION: Our findings demonstrate the capacity of PAR-2 activation to augment TGFß production and promote hepatic fibrosis in mice and to induce a profibrogenic phenotype in human HSCs. PAR-2 antagonists have recently been developed and may represent a novel therapeutic approach in preventing fibrosis in patients with chronic liver disease.


Subject(s)
Carbon Tetrachloride/adverse effects , Disease Progression , Hepatic Stellate Cells/physiology , Liver Cirrhosis/chemically induced , Liver Cirrhosis/physiopathology , Receptor, PAR-2/physiology , Animals , Cell Proliferation/drug effects , Cells, Cultured , Collagen/metabolism , Cytokines/metabolism , Disease Models, Animal , Hepatic Stellate Cells/cytology , Humans , Hydroxyproline/metabolism , In Vitro Techniques , Liver/metabolism , Liver/pathology , Male , Matrix Metalloproteinase 2/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligopeptides/pharmacology , Receptor, PAR-2/agonists , Receptor, PAR-2/deficiency , Tissue Inhibitor of Metalloproteinase-1/metabolism , Transforming Growth Factor beta/metabolism
6.
Cell Transplant ; 19(9): 1157-68, 2010.
Article in English | MEDLINE | ID: mdl-20447339

ABSTRACT

Chronic liver injury and inflammation lead to hepatic fibrosis, cirrhosis, and liver failure. Embryonic and mesenchymal stem cells have been shown to reduce experimental liver fibrosis but have potential limitations, including the formation of dysplastic precursors, tumors, and profibrogenic cells. Other stem-like cells may reduce hepatic inflammation and fibrosis without tumor and profibrogenic cell formation. To test this hypothesis we transplanted human amnion epithelial cells (hAEC), isolated from term delivered placenta, into immunocompetent C57/BL6 mice at week 2 of a 4-week regimen of carbon tetrachloride (CCl4) exposure to induce liver fibrosis. Two weeks following hAEC infusion, intact cells expressing the human-specific markers inner mitochondrial membrane protein and human leukocyte antigen-G were found in mouse liver without evidence of host rejection of the transplanted cells. Human albumin, known to be produced by hAEC, was detected in sera of hAEC-treated mice. Human DNA was detected in mouse liver and also spleen, lungs, and heart of some animals. Following hAEC transplantation, CCl4-treated animals showed decreased serum ALT levels and reduced hepatocyte apoptosis, compared to controls. hAEC-treated mouse liver had lower TNF-α and IL-6 protein levels and higher IL-10 compared to animals given CCl4 alone. Compared to CCl4 controls, hAEC-treated mice showed fewer activated collagen-producing hepatic stellate cells and less fibrosis area and collagen content. Reduced hepatic TGF-ß levels in conjunction with a twofold increase in the active form of the collagen-degrading enzyme matrix metalloproteinase-2 in hAEC-treated mice compared to CCl4 controls may account for the reduction in fibrosis. hAEC transplantation into immunocompetent mice leads to cell engraftment, reduced hepatocyte apoptosis, and decreased hepatic inflammation and fibrosis.


Subject(s)
Amnion/cytology , Cell Transplantation/methods , Liver Cirrhosis/surgery , Animals , Apoptosis/physiology , Carbon Tetrachloride , Carbon Tetrachloride Poisoning/immunology , Carbon Tetrachloride Poisoning/pathology , Carbon Tetrachloride Poisoning/surgery , Cell Separation/methods , Epithelial Cells/cytology , Female , Humans , Immunohistochemistry , Liver/drug effects , Liver/metabolism , Liver/pathology , Liver Cirrhosis/chemically induced , Liver Cirrhosis/immunology , Liver Cirrhosis/pathology , Male , Mice , Mice, Inbred C57BL , Pregnancy , Transforming Growth Factor beta/metabolism , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...