Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Type of study
Language
Publication year range
1.
PLoS One ; 14(7): e0215557, 2019.
Article in English | MEDLINE | ID: mdl-31291257

ABSTRACT

BACKGROUND: Chronic inflammation is the driver of liver injury and results in progressive fibrosis and eventual cirrhosis with consequences including both liver failure and liver cancer. We have previously described increased expression of the highly multifunctional glycoprotein CD147 in liver injury. This work describes a novel role of CD147 in liver inflammation and the importance of leukocyte aggregates in determining the extent of liver injury. METHODS: Non-diseased, progressive injury, and cirrhotic liver from humans and mice were examined using a mAb targeting CD147. Inflammatory cell subsets were assessed by multiparameter flow cytometry. RESULTS: In liver injury, we observe abundant, intrahepatic leukocyte clusters defined as ≥5 adjacent CD45+ cells which we have termed "leukocyte aggregates". We have shown that these leukocyte aggregates have a significant effect in determining the extent of liver injury. If CD147 is blocked in vivo, these leukocyte aggregates diminish in size and number, together with a marked significant reduction in liver injury including fibrosis. This is accompanied by no change in overall intrahepatic leukocyte numbers. Further, blocking of aggregation formation occurs prior to an appreciable increase in inflammatory markers or fibrosis. Additionally, there were no observed, "off-target" or unpredicted effects in targeting CD147. CONCLUSION: CD147 mediates leukocyte aggregation which is associated with the development of liver injury. This is not a secondary effect, but a cause of injury as aggregate formation proceeds other markers of injury. Leukocyte aggregation has been previously described in inflammation dating back over many decades. Here we demonstrate that leukocyte aggregates determine the extent of liver injury.


Subject(s)
Basigin/metabolism , Leukocytes/immunology , Liver/immunology , Liver/injuries , Animals , Basigin/genetics , Cell Aggregation/immunology , Hepatocytes/immunology , Hepatocytes/pathology , Humans , Leukocytes/classification , Leukocytes/pathology , Liver/pathology , Liver Cirrhosis/genetics , Liver Cirrhosis/immunology , Liver Cirrhosis/pathology , Liver Cirrhosis, Experimental/genetics , Liver Cirrhosis, Experimental/immunology , Liver Cirrhosis, Experimental/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Up-Regulation
2.
J Gastroenterol Hepatol ; 30(12): 1696-704, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26239824

ABSTRACT

Chronic liver disease causes significant morbidity and mortality through progressive fibrosis, cirrhosis, and liver cancer. The classical theory of fibrogenesis has hepatic stellate cells (HSCs) as the principal and only significant source of abnormal extracellular matrix (ECM). Further, HSCs have the major role in abnormal ECM turnover. It is the death of hepatocytes, as the initial target of injury, that initiates a sequence of events including the recruitment of inflammatory cells and activation of HSCs. Following this initial response, the ongoing insult to hepatocytes is regarded as perpetuating injury, but otherwise, hepatocytes are regarded as "victims" and "bystanders" in progressive fibrosis. Recent developments, however, challenge this view and suggest the concept of the hepatocyte being an active participant in liver injury. It is clear now that hepatocytes undergo phenotypic changes, adapt to injury, and react to the altered microenvironment. In this review, we describe studies showing that hepatocytes contribute to progressive fibrosis by direct manipulation of the surrounding ECM and through signaling to effector cells, particularly HSCs and intrahepatic immune cells. Together, these findings suggest an active "accomplice" role for the hepatocyte in progressive liver fibrosis and highlight novel pathways that could be targeted for development of future anti-fibrotic therapies.


Subject(s)
Hepatocytes/physiology , Liver Diseases/etiology , Cell Death , Disease Progression , Extracellular Matrix , Hepatocytes/immunology , Hepatocytes/pathology , Humans , Liver Cirrhosis/etiology , Liver Cirrhosis/immunology , Liver Cirrhosis/pathology , Liver Diseases/immunology , Liver Diseases/pathology , Liver Regeneration
3.
PLoS One ; 9(7): e90571, 2014.
Article in English | MEDLINE | ID: mdl-25076423

ABSTRACT

BACKGROUND: The classical paradigm of liver injury asserts that hepatic stellate cells (HSC) produce, remodel and turnover the abnormal extracellular matrix (ECM) of fibrosis via matrix metalloproteinases (MMPs). In extrahepatic tissues MMP production is regulated by a number of mechanisms including expression of the glycoprotein CD147. Previously, we have shown that CD147 is expressed on hepatocytes but not within the fibrotic septa in cirrhosis [1]. Therefore, we investigated if hepatocytes produce MMPs, regulated by CD147, which are capable of remodelling fibrotic ECM independent of the HSC. METHODS: Non-diseased, fibrotic and cirrhotic livers were examined for MMP activity and markers of fibrosis in humans and mice. CD147 expression and MMP activity were co-localised by in-situ zymography. The role of CD147 was studied in-vitro with siRNA to CD147 in hepatocytes and in-vivo in mice with CCl4 induced liver injury using ãCD147 antibody intervention. RESULTS: In liver fibrosis in both human and mouse tissue MMP expression and activity (MMP-2, -9, -13 and -14) increased with progressive injury and localised to hepatocytes. Additionally, as expected, MMPs were abundantly expressed by activated HSC. Further, with progressive fibrosis there was expression of CD147, which localised to hepatocytes but not to HSC. Functionally significant in-vitro regulation of hepatocyte MMP production by CD147 was demonstrated using siRNA to CD147 that decreased hepatocyte MMP-2 and -9 expression/activity. Further, in-vivo α-CD147 antibody intervention decreased liver MMP-2, -9, -13, -14, TGF-ß and α-SMA expression in CCl4 treated mice compared to controls. CONCLUSION: We have shown that hepatocytes produce active MMPs and that the glycoprotein CD147 regulates hepatocyte MMP expression. Targeting CD147 regulates hepatocyte MMP production both in-vitro and in-vivo, with the net result being reduced fibrotic matrix turnover in-vivo. Therefore, CD147 regulation of hepatocyte MMP is a novel pathway that could be targeted by future anti-fibrogenic agents.


Subject(s)
Basigin/metabolism , Hepatocytes/metabolism , Liver Cirrhosis/pathology , Matrix Metalloproteinases/metabolism , Animals , Basigin/chemistry , Basigin/genetics , Cells, Cultured , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/metabolism , Hepatocytes/cytology , Humans , Hydrocarbons, Brominated/toxicity , Immunohistochemistry , Liver/metabolism , Liver/pathology , Liver Cirrhosis/metabolism , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinases/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Polymerase Chain Reaction , RNA Interference , RNA, Small Interfering/metabolism , Tissue Inhibitor of Metalloproteinases/metabolism
4.
J Hepatol ; 60(1): 143-51, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23978713

ABSTRACT

BACKGROUND & AIMS: In vertebrates, canonical Hedgehog (Hh) pathway activation requires Smoothened (SMO) translocation to the primary cilium (Pc), followed by a GLI-mediated transcriptional response. In addition, a similar gene regulation occurs in response to growth factors/cytokines, although independently of SMO signalling. The Hh pathway plays a critical role in liver fibrosis/regeneration, however, the mechanism of activation in chronic liver injury is poorly understood. This study aimed to characterise Hh pathway activation upon thioacetamide (TAA)-induced chronic liver injury in vivo by defining Hh-responsive cells, namely cells harbouring Pc and Pc-localised SMO. METHODS: C57BL/6 mice (wild-type or Ptc1(+/-)) were TAA-treated. Liver injury and Hh ligand/pathway mRNA and protein expression were assessed in vivo. SMO/GLI manipulation and SMO-dependent/independent activation of GLI-mediated transcriptional response in Pc-positive (Pc(+)) cells were studied in vitro. RESULTS: In vivo, Hh activation was progressively induced following TAA. At the epithelial-mesenchymal interface, injured hepatocytes produced Hh ligands. Progenitors, myofibroblasts, leukocytes and hepatocytes were GLI2(+). Pc(+) cells increased following TAA, but only EpCAM(+)/GLI2(+) progenitors were Pc(+)/SMO(+). In vitro, SMO knockdown/hGli3-R overexpression reduced proliferation/viability in Pc(+) progenitors, whilst increased proliferation occurred with hGli1 overexpression. HGF induced GLI transcriptional activity independently of Pc/SMO. Ptc1(+/-) mice exhibited increased progenitor, myofibroblast and fibrosis responses. CONCLUSIONS: In chronic liver injury, Pc(+) progenitors receive Hh ligand signals and process it through Pc/SMO-dependent activation of GLI-mediated transcriptional response. Pc/SMO-independent GLI activation likely occurs in Pc(-)/GLI2(+) cells. Increased fibrosis in Hh gain-of-function mice likely occurs by primary progenitor expansion/proliferation and secondary fibrotic myofibroblast expansion, in close contact with progenitors.


Subject(s)
Chemical and Drug Induced Liver Injury/pathology , Cilia/physiology , Hedgehog Proteins/physiology , Liver/pathology , Signal Transduction/physiology , Animals , Chronic Disease , Epithelial-Mesenchymal Transition , Kruppel-Like Transcription Factors/analysis , Kruppel-Like Transcription Factors/physiology , Male , Mice , Mice, Inbred C57BL , Nuclear Proteins/analysis , Receptors, G-Protein-Coupled/physiology , Smoothened Receptor , Thioacetamide , Zinc Finger Protein GLI1 , Zinc Finger Protein Gli2
SELECTION OF CITATIONS
SEARCH DETAIL
...