Cargando…

Disruption of the Smad7 gene enhances CCI(4)-dependent liver damage and fibrogenesis in mice

Transforming growth factor-β (TGF-β) signalling is induced in liver as a consequence of damage and contributes to wound healing with transient activation, whereas it mediates fibrogenesis with long-term up-regulation in chronic disease. Smad-dependent TGF-β effects are blunted by antagonistic Smad7,...

Descripción completa

Detalles Bibliográficos
Autores principales: Hamzavi, Jafar, Ehnert, Sabrina, Godoy, Patricio, Ciuclan, Loredana, Weng, Honglei, Mertens, Peter R, Heuchel, Rainer, Dooley, Steven
Formato: Online Artículo Texto
Lenguaje:English
Publicado: John Wiley & Sons, Ltd 2008
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4506177/
https://www.ncbi.nlm.nih.gov/pubmed/18266971
http://dx.doi.org/10.1111/j.1582-4934.2008.00262.x
_version_ 1782381661161259008
author Hamzavi, Jafar
Ehnert, Sabrina
Godoy, Patricio
Ciuclan, Loredana
Weng, Honglei
Mertens, Peter R
Heuchel, Rainer
Dooley, Steven
author_facet Hamzavi, Jafar
Ehnert, Sabrina
Godoy, Patricio
Ciuclan, Loredana
Weng, Honglei
Mertens, Peter R
Heuchel, Rainer
Dooley, Steven
author_sort Hamzavi, Jafar
collection PubMed
description Transforming growth factor-β (TGF-β) signalling is induced in liver as a consequence of damage and contributes to wound healing with transient activation, whereas it mediates fibrogenesis with long-term up-regulation in chronic disease. Smad-dependent TGF-β effects are blunted by antagonistic Smad7, which is transcriptionally activated as an immediate early response upon initiation of TGF-β signalling in most cell types, thereby providing negative feedback regulation. Smad7 can be induced by other cytokines, e.g. IFN-γ, leading to a crosstalk of these signalling pathways. Here we report on a novel mouse strain, denoted S7ΔE1, with a deletion of exon I from the endogenous smad7 gene. The mice were viable and exhibited normal adult liver architecture. To obtain insight into Smad7-depend-ent protective effects, chronic liver damage was induced in mice by carbon tetrachloride (CCI(4)) administration. Subsequent treatment, elevated serum liver enzymes indicated enhanced liver damage in mice lacking functional Smad7. CCI(4)-dependent Smad2 phosphoryla-tion was pronounced in S7ΔE1 mice and accompanied by increased numbers of α-smooth muscle actin positive ‘activated’ HSCs. There was evidence for matrix accumulation, with elevated collagen deposition as assessed morphometrically in Sirius red stained tissue and confirmed with higher levels of hydroxyproline in S7ΔE1 mice. In addition, the number of CD43 positive infiltrating lymphocytes as well as of apoptotic hepatocytes was increased. Studies with primary hepatocytes from S7ΔE1 and wild-type mice indicate that in the absence of functional Smad7 protein, hepatocytes are more sensitive for TGF-β effects resulting in enhanced cell death. Furthermore, S7ΔE1 hepatocytes display increased oxidative stress and cell damage in response to CCI(4), as measured by reactive oxygen species production, glutathione depletion, lactate dehydrogenase release and lipid peroxidation. Using an ALK-5 inhibitor all investigated CCI(4) effects on hepatocytes were blunted, confirming participation of TGF-β signalling. We conclude that Smad7 mediates a protective effect from adverse TGF-β signalling in damaged liver, re-iterating its negative regulatory loop on signalling.
format Online
Article
Text
id pubmed-4506177
institution National Center for Biotechnology Information
language English
publishDate 2008
publisher John Wiley & Sons, Ltd
record_format MEDLINE/PubMed
spelling pubmed-45061772015-07-22 Disruption of the Smad7 gene enhances CCI(4)-dependent liver damage and fibrogenesis in mice Hamzavi, Jafar Ehnert, Sabrina Godoy, Patricio Ciuclan, Loredana Weng, Honglei Mertens, Peter R Heuchel, Rainer Dooley, Steven J Cell Mol Med Articles Transforming growth factor-β (TGF-β) signalling is induced in liver as a consequence of damage and contributes to wound healing with transient activation, whereas it mediates fibrogenesis with long-term up-regulation in chronic disease. Smad-dependent TGF-β effects are blunted by antagonistic Smad7, which is transcriptionally activated as an immediate early response upon initiation of TGF-β signalling in most cell types, thereby providing negative feedback regulation. Smad7 can be induced by other cytokines, e.g. IFN-γ, leading to a crosstalk of these signalling pathways. Here we report on a novel mouse strain, denoted S7ΔE1, with a deletion of exon I from the endogenous smad7 gene. The mice were viable and exhibited normal adult liver architecture. To obtain insight into Smad7-depend-ent protective effects, chronic liver damage was induced in mice by carbon tetrachloride (CCI(4)) administration. Subsequent treatment, elevated serum liver enzymes indicated enhanced liver damage in mice lacking functional Smad7. CCI(4)-dependent Smad2 phosphoryla-tion was pronounced in S7ΔE1 mice and accompanied by increased numbers of α-smooth muscle actin positive ‘activated’ HSCs. There was evidence for matrix accumulation, with elevated collagen deposition as assessed morphometrically in Sirius red stained tissue and confirmed with higher levels of hydroxyproline in S7ΔE1 mice. In addition, the number of CD43 positive infiltrating lymphocytes as well as of apoptotic hepatocytes was increased. Studies with primary hepatocytes from S7ΔE1 and wild-type mice indicate that in the absence of functional Smad7 protein, hepatocytes are more sensitive for TGF-β effects resulting in enhanced cell death. Furthermore, S7ΔE1 hepatocytes display increased oxidative stress and cell damage in response to CCI(4), as measured by reactive oxygen species production, glutathione depletion, lactate dehydrogenase release and lipid peroxidation. Using an ALK-5 inhibitor all investigated CCI(4) effects on hepatocytes were blunted, confirming participation of TGF-β signalling. We conclude that Smad7 mediates a protective effect from adverse TGF-β signalling in damaged liver, re-iterating its negative regulatory loop on signalling. John Wiley & Sons, Ltd 2008-10 2008-02-06 /pmc/articles/PMC4506177/ /pubmed/18266971 http://dx.doi.org/10.1111/j.1582-4934.2008.00262.x Text en © 2008 The Authors Journal compilation © 2008 Foundation for Cellular and Molecular Medicine/Blackwell Publishing Ltd
spellingShingle Articles
Hamzavi, Jafar
Ehnert, Sabrina
Godoy, Patricio
Ciuclan, Loredana
Weng, Honglei
Mertens, Peter R
Heuchel, Rainer
Dooley, Steven
Disruption of the Smad7 gene enhances CCI(4)-dependent liver damage and fibrogenesis in mice
title Disruption of the Smad7 gene enhances CCI(4)-dependent liver damage and fibrogenesis in mice
title_full Disruption of the Smad7 gene enhances CCI(4)-dependent liver damage and fibrogenesis in mice
title_fullStr Disruption of the Smad7 gene enhances CCI(4)-dependent liver damage and fibrogenesis in mice
title_full_unstemmed Disruption of the Smad7 gene enhances CCI(4)-dependent liver damage and fibrogenesis in mice
title_short Disruption of the Smad7 gene enhances CCI(4)-dependent liver damage and fibrogenesis in mice
title_sort disruption of the smad7 gene enhances cci(4)-dependent liver damage and fibrogenesis in mice
topic Articles
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4506177/
https://www.ncbi.nlm.nih.gov/pubmed/18266971
http://dx.doi.org/10.1111/j.1582-4934.2008.00262.x
work_keys_str_mv AT hamzavijafar disruptionofthesmad7geneenhancescci4dependentliverdamageandfibrogenesisinmice
AT ehnertsabrina disruptionofthesmad7geneenhancescci4dependentliverdamageandfibrogenesisinmice
AT godoypatricio disruptionofthesmad7geneenhancescci4dependentliverdamageandfibrogenesisinmice
AT ciuclanloredana disruptionofthesmad7geneenhancescci4dependentliverdamageandfibrogenesisinmice
AT wenghonglei disruptionofthesmad7geneenhancescci4dependentliverdamageandfibrogenesisinmice
AT mertenspeterr disruptionofthesmad7geneenhancescci4dependentliverdamageandfibrogenesisinmice
AT heuchelrainer disruptionofthesmad7geneenhancescci4dependentliverdamageandfibrogenesisinmice
AT dooleysteven disruptionofthesmad7geneenhancescci4dependentliverdamageandfibrogenesisinmice