PPAR-gamma activation is associated with reduced liver ischemia-reperfusion injury and altered tissue-resident macrophages polarization in a mouse model.

PPAR-gamma (γ) is highly expressed in macrophages and its activation affects their polarization. The effect of PPAR-γ activation on Kupffer cells (KCs) and liver ischemia-reperfusion injury (IRI) has not yet been evaluated. We investigated the effect of PPAR-γ activation on KC-polarization and IRI.S...

Full description

Bibliographic Details
Main Authors: Ivan Linares, Kaveh Farrokhi, Juan Echeverri, Johan Moritz Kaths, Dagmar Kollmann, Matyas Hamar, Peter Urbanellis, Sujani Ganesh, Oyedele A Adeyi, Paul Yip, Markus Selzner, Nazia Selzner
Format: Article
Language:English
Published: Public Library of Science (PLoS) 2018-01-01
Series:PLoS ONE
Online Access:http://europepmc.org/articles/PMC5884549?pdf=render
_version_ 1811233194917232640
author Ivan Linares
Kaveh Farrokhi
Juan Echeverri
Johan Moritz Kaths
Dagmar Kollmann
Matyas Hamar
Peter Urbanellis
Sujani Ganesh
Oyedele A Adeyi
Paul Yip
Markus Selzner
Nazia Selzner
author_facet Ivan Linares
Kaveh Farrokhi
Juan Echeverri
Johan Moritz Kaths
Dagmar Kollmann
Matyas Hamar
Peter Urbanellis
Sujani Ganesh
Oyedele A Adeyi
Paul Yip
Markus Selzner
Nazia Selzner
author_sort Ivan Linares
collection DOAJ
description PPAR-gamma (γ) is highly expressed in macrophages and its activation affects their polarization. The effect of PPAR-γ activation on Kupffer cells (KCs) and liver ischemia-reperfusion injury (IRI) has not yet been evaluated. We investigated the effect of PPAR-γ activation on KC-polarization and IRI.Seventy percent (70%) liver ischemia was induced for 60mins. PPAR-γ-agonist or vehicle was administrated before reperfusion. PPAR-γ-antagonist was used to block PPAR-γ activation. Liver injury, necrosis, and apoptosis were assessed post-reperfusion. Flow-cytometry determined KC-phenotypes (pro-inflammatory Nitric Oxide +, anti-inflammatory CD206+ and anti-inflammatory IL-10+).Liver injury assessed by serum AST was significantly decreased in PPAR-γ-agonist versus control group at all time points post reperfusion (1hr: 3092±105 vs 4469±551; p = 0.042; 6hr: 7041±1160 vs 12193±1143; p = 0.015; 12hr: 5746±328 vs 8608±1259; p = 0.049). Furthermore, liver apoptosis measured by TUNEL-staining was significantly reduced in PPAR-γ-agonist versus control group post reperfusion (1hr:2.46±0.49 vs 6.90±0.85%;p = 0.001; 6hr:26.40±2.93 vs 50.13±8.29%; p = 0.048). H&E staining demonstrated less necrosis in PPAR-γ-agonist versus control group (24hr:26.66±4.78 vs 45.62±4.57%; p = 0.032). The percentage of pro-inflammatory NO+ KCs was significantly lower at all post reperfusion time points in the PPAR-γ-agonist versus control group (1hr:28.49±4.99 vs 53.54±9.15%; p = 0.040; 6hr:5.51±0.54 vs 31.12±9.58%; p = 0.009; 24hr:4.15±1.50 vs 17.10±4.77%; p = 0.043). In contrast, percentage of anti-inflammatory CD206+ KCs was significantly higher in PPAR-γ-agonist versus control group prior to IRI (8.62±0.96 vs 4.88 ±0.50%; p = 0.04). Administration of PPAR-γ-antagonist reversed the beneficial effects on AST, apoptosis, and pro-inflammatory NO+ KCs.PPAR-γ activation reduces IRI and decreases the pro-inflammatory NO+ Kupffer cells. PPAR-γ activation can become an important tool to improve outcomes in liver surgery through decreasing the pro-inflammatory phenotype of KCs and IRI.
first_indexed 2024-04-12T11:17:35Z
format Article
id doaj.art-1f548920ba954917acb3fc7ec850a94d
institution Directory Open Access Journal
issn 1932-6203
language English
last_indexed 2024-04-12T11:17:35Z
publishDate 2018-01-01
publisher Public Library of Science (PLoS)
record_format Article
series PLoS ONE
spelling doaj.art-1f548920ba954917acb3fc7ec850a94d2022-12-22T03:35:28ZengPublic Library of Science (PLoS)PLoS ONE1932-62032018-01-01134e019521210.1371/journal.pone.0195212PPAR-gamma activation is associated with reduced liver ischemia-reperfusion injury and altered tissue-resident macrophages polarization in a mouse model.Ivan LinaresKaveh FarrokhiJuan EcheverriJohan Moritz KathsDagmar KollmannMatyas HamarPeter UrbanellisSujani GaneshOyedele A AdeyiPaul YipMarkus SelznerNazia SelznerPPAR-gamma (γ) is highly expressed in macrophages and its activation affects their polarization. The effect of PPAR-γ activation on Kupffer cells (KCs) and liver ischemia-reperfusion injury (IRI) has not yet been evaluated. We investigated the effect of PPAR-γ activation on KC-polarization and IRI.Seventy percent (70%) liver ischemia was induced for 60mins. PPAR-γ-agonist or vehicle was administrated before reperfusion. PPAR-γ-antagonist was used to block PPAR-γ activation. Liver injury, necrosis, and apoptosis were assessed post-reperfusion. Flow-cytometry determined KC-phenotypes (pro-inflammatory Nitric Oxide +, anti-inflammatory CD206+ and anti-inflammatory IL-10+).Liver injury assessed by serum AST was significantly decreased in PPAR-γ-agonist versus control group at all time points post reperfusion (1hr: 3092±105 vs 4469±551; p = 0.042; 6hr: 7041±1160 vs 12193±1143; p = 0.015; 12hr: 5746±328 vs 8608±1259; p = 0.049). Furthermore, liver apoptosis measured by TUNEL-staining was significantly reduced in PPAR-γ-agonist versus control group post reperfusion (1hr:2.46±0.49 vs 6.90±0.85%;p = 0.001; 6hr:26.40±2.93 vs 50.13±8.29%; p = 0.048). H&E staining demonstrated less necrosis in PPAR-γ-agonist versus control group (24hr:26.66±4.78 vs 45.62±4.57%; p = 0.032). The percentage of pro-inflammatory NO+ KCs was significantly lower at all post reperfusion time points in the PPAR-γ-agonist versus control group (1hr:28.49±4.99 vs 53.54±9.15%; p = 0.040; 6hr:5.51±0.54 vs 31.12±9.58%; p = 0.009; 24hr:4.15±1.50 vs 17.10±4.77%; p = 0.043). In contrast, percentage of anti-inflammatory CD206+ KCs was significantly higher in PPAR-γ-agonist versus control group prior to IRI (8.62±0.96 vs 4.88 ±0.50%; p = 0.04). Administration of PPAR-γ-antagonist reversed the beneficial effects on AST, apoptosis, and pro-inflammatory NO+ KCs.PPAR-γ activation reduces IRI and decreases the pro-inflammatory NO+ Kupffer cells. PPAR-γ activation can become an important tool to improve outcomes in liver surgery through decreasing the pro-inflammatory phenotype of KCs and IRI.http://europepmc.org/articles/PMC5884549?pdf=render
spellingShingle Ivan Linares
Kaveh Farrokhi
Juan Echeverri
Johan Moritz Kaths
Dagmar Kollmann
Matyas Hamar
Peter Urbanellis
Sujani Ganesh
Oyedele A Adeyi
Paul Yip
Markus Selzner
Nazia Selzner
PPAR-gamma activation is associated with reduced liver ischemia-reperfusion injury and altered tissue-resident macrophages polarization in a mouse model.
PLoS ONE
title PPAR-gamma activation is associated with reduced liver ischemia-reperfusion injury and altered tissue-resident macrophages polarization in a mouse model.
title_full PPAR-gamma activation is associated with reduced liver ischemia-reperfusion injury and altered tissue-resident macrophages polarization in a mouse model.
title_fullStr PPAR-gamma activation is associated with reduced liver ischemia-reperfusion injury and altered tissue-resident macrophages polarization in a mouse model.
title_full_unstemmed PPAR-gamma activation is associated with reduced liver ischemia-reperfusion injury and altered tissue-resident macrophages polarization in a mouse model.
title_short PPAR-gamma activation is associated with reduced liver ischemia-reperfusion injury and altered tissue-resident macrophages polarization in a mouse model.
title_sort ppar gamma activation is associated with reduced liver ischemia reperfusion injury and altered tissue resident macrophages polarization in a mouse model
url http://europepmc.org/articles/PMC5884549?pdf=render
work_keys_str_mv AT ivanlinares ppargammaactivationisassociatedwithreducedliverischemiareperfusioninjuryandalteredtissueresidentmacrophagespolarizationinamousemodel
AT kavehfarrokhi ppargammaactivationisassociatedwithreducedliverischemiareperfusioninjuryandalteredtissueresidentmacrophagespolarizationinamousemodel
AT juanecheverri ppargammaactivationisassociatedwithreducedliverischemiareperfusioninjuryandalteredtissueresidentmacrophagespolarizationinamousemodel
AT johanmoritzkaths ppargammaactivationisassociatedwithreducedliverischemiareperfusioninjuryandalteredtissueresidentmacrophagespolarizationinamousemodel
AT dagmarkollmann ppargammaactivationisassociatedwithreducedliverischemiareperfusioninjuryandalteredtissueresidentmacrophagespolarizationinamousemodel
AT matyashamar ppargammaactivationisassociatedwithreducedliverischemiareperfusioninjuryandalteredtissueresidentmacrophagespolarizationinamousemodel
AT peterurbanellis ppargammaactivationisassociatedwithreducedliverischemiareperfusioninjuryandalteredtissueresidentmacrophagespolarizationinamousemodel
AT sujaniganesh ppargammaactivationisassociatedwithreducedliverischemiareperfusioninjuryandalteredtissueresidentmacrophagespolarizationinamousemodel
AT oyedeleaadeyi ppargammaactivationisassociatedwithreducedliverischemiareperfusioninjuryandalteredtissueresidentmacrophagespolarizationinamousemodel
AT paulyip ppargammaactivationisassociatedwithreducedliverischemiareperfusioninjuryandalteredtissueresidentmacrophagespolarizationinamousemodel
AT markusselzner ppargammaactivationisassociatedwithreducedliverischemiareperfusioninjuryandalteredtissueresidentmacrophagespolarizationinamousemodel
AT naziaselzner ppargammaactivationisassociatedwithreducedliverischemiareperfusioninjuryandalteredtissueresidentmacrophagespolarizationinamousemodel