Therapeutic Targeting of Myeloperoxidase Attenuates NASH in Mice

Myeloperoxidase (MPO) activity has been associated with the metabolic syndrome, cardiovascular and liver disease. Here, we evaluate the therapeutic potential of MPO inhibition on nonalcoholic steatohepatitis (NASH) and NASH‐induced fibrosis, the main determinant of outcomes. MPO plasma levels were e...

Full description

Bibliographic Details
Main Authors: Anja Christina Koop, Nina Doreen Thiele, David Steins, Erik Michaëlsson, Malte Wehmeyer, Ludger Scheja, Babett Steglich, Samuel Huber, Julian Schulze zur Wiesch, Ansgar W. Lohse, Jörg Heeren, Johannes Kluwe
Format: Article
Language:English
Published: Wiley 2020-10-01
Series:Hepatology Communications
Online Access:https://doi.org/10.1002/hep4.1566
id doaj-5f95b267bde54b54b722eb920d066cb8
record_format Article
spelling doaj-5f95b267bde54b54b722eb920d066cb82021-02-24T07:56:04ZengWileyHepatology Communications2471-254X2020-10-014101441145810.1002/hep4.1566Therapeutic Targeting of Myeloperoxidase Attenuates NASH in MiceAnja Christina Koop0Nina Doreen Thiele1David Steins2Erik Michaëlsson3Malte Wehmeyer4Ludger Scheja5Babett Steglich6Samuel Huber7Julian Schulze zur Wiesch8Ansgar W. Lohse9Jörg Heeren10Johannes Kluwe111st Department of Medicine University Medical Center Hamburg‐Eppendorf Hamburg Germany1st Department of Medicine University Medical Center Hamburg‐Eppendorf Hamburg Germany1st Department of Medicine University Medical Center Hamburg‐Eppendorf Hamburg GermanyBioscience Cardiovascular, Research and Early Development Cardiovascular, Renal and Metabolism BioPharmaceuticals R&D AstraZeneca Gothenburg Sweden1st Department of Medicine University Medical Center Hamburg‐Eppendorf Hamburg GermanyDepartment of Biochemistry and Molecular Cell Biology University Medical Center Hamburg‐Eppendorf Hamburg Germany1st Department of Medicine University Medical Center Hamburg‐Eppendorf Hamburg Germany1st Department of Medicine University Medical Center Hamburg‐Eppendorf Hamburg Germany1st Department of Medicine University Medical Center Hamburg‐Eppendorf Hamburg Germany1st Department of Medicine University Medical Center Hamburg‐Eppendorf Hamburg GermanyDepartment of Biochemistry and Molecular Cell Biology University Medical Center Hamburg‐Eppendorf Hamburg Germany1st Department of Medicine University Medical Center Hamburg‐Eppendorf Hamburg GermanyMyeloperoxidase (MPO) activity has been associated with the metabolic syndrome, cardiovascular and liver disease. Here, we evaluate the therapeutic potential of MPO inhibition on nonalcoholic steatohepatitis (NASH) and NASH‐induced fibrosis, the main determinant of outcomes. MPO plasma levels were elevated in patients with nonalcoholic fatty liver disease (NAFLD) compared with healthy controls. In a second cohort, hepatic MPO messenger RNA expression correlated with higher body mass index and hemoglobin A1c, both being risk factors for NAFLD. We could establish by immunohistochemistry that MPO‐positive cells were recruited to the liver in various mouse models of fibrogenic liver injury, including bile duct ligation, carbon tetrachloride (CCl4) treatment, spontaneous liver fibrogenesis in multidrug resistance 2 knockout (MDR2 KO) mice, and NASH‐inducing diet. Comparison of MPO‐deficient mice and their wild‐type littermates exposed to a high‐caloric diet revealed that MPO deficiency protects against NASH‐related liver injury and fibrosis. In line with this, hepatic gene expression analysis demonstrated a MPO‐dependent activation of pathways relevant for wound healing, inflammation, and cell death in NASH. MPO deficiency did not affect NAFLD‐independent liver injury and fibrosis in MDR2 KO or CCl4‐treated mice. Finally, we treated wild‐type mice exposed to NASH‐inducing diet with an oral MPO inhibitor. Pharmacological MPO inhibition not only reduced markers of MPO‐mediated liver damage, serum alanine aminotransferase levels, and hepatic steatosis, but also significantly decreased NASH‐induced liver fibrosis. MPO inhibitor treatment, but not MPO deficiency, significantly altered gut microbiota including a significant expansion of Akkermansia muciniphila. Conclusions: MPO specifically promotes NASH‐induced liver fibrosis. Pharmacological MPO inhibition attenuates NASH progression and NASH‐induced liver fibrosis in mice and is associated with beneficial changes of intestinal microbiota.https://doi.org/10.1002/hep4.1566
collection DOAJ
language English
format Article
sources DOAJ
author Anja Christina Koop
Nina Doreen Thiele
David Steins
Erik Michaëlsson
Malte Wehmeyer
Ludger Scheja
Babett Steglich
Samuel Huber
Julian Schulze zur Wiesch
Ansgar W. Lohse
Jörg Heeren
Johannes Kluwe
spellingShingle Anja Christina Koop
Nina Doreen Thiele
David Steins
Erik Michaëlsson
Malte Wehmeyer
Ludger Scheja
Babett Steglich
Samuel Huber
Julian Schulze zur Wiesch
Ansgar W. Lohse
Jörg Heeren
Johannes Kluwe
Therapeutic Targeting of Myeloperoxidase Attenuates NASH in Mice
Hepatology Communications
author_facet Anja Christina Koop
Nina Doreen Thiele
David Steins
Erik Michaëlsson
Malte Wehmeyer
Ludger Scheja
Babett Steglich
Samuel Huber
Julian Schulze zur Wiesch
Ansgar W. Lohse
Jörg Heeren
Johannes Kluwe
author_sort Anja Christina Koop
title Therapeutic Targeting of Myeloperoxidase Attenuates NASH in Mice
title_short Therapeutic Targeting of Myeloperoxidase Attenuates NASH in Mice
title_full Therapeutic Targeting of Myeloperoxidase Attenuates NASH in Mice
title_fullStr Therapeutic Targeting of Myeloperoxidase Attenuates NASH in Mice
title_full_unstemmed Therapeutic Targeting of Myeloperoxidase Attenuates NASH in Mice
title_sort therapeutic targeting of myeloperoxidase attenuates nash in mice
publisher Wiley
series Hepatology Communications
issn 2471-254X
publishDate 2020-10-01
description Myeloperoxidase (MPO) activity has been associated with the metabolic syndrome, cardiovascular and liver disease. Here, we evaluate the therapeutic potential of MPO inhibition on nonalcoholic steatohepatitis (NASH) and NASH‐induced fibrosis, the main determinant of outcomes. MPO plasma levels were elevated in patients with nonalcoholic fatty liver disease (NAFLD) compared with healthy controls. In a second cohort, hepatic MPO messenger RNA expression correlated with higher body mass index and hemoglobin A1c, both being risk factors for NAFLD. We could establish by immunohistochemistry that MPO‐positive cells were recruited to the liver in various mouse models of fibrogenic liver injury, including bile duct ligation, carbon tetrachloride (CCl4) treatment, spontaneous liver fibrogenesis in multidrug resistance 2 knockout (MDR2 KO) mice, and NASH‐inducing diet. Comparison of MPO‐deficient mice and their wild‐type littermates exposed to a high‐caloric diet revealed that MPO deficiency protects against NASH‐related liver injury and fibrosis. In line with this, hepatic gene expression analysis demonstrated a MPO‐dependent activation of pathways relevant for wound healing, inflammation, and cell death in NASH. MPO deficiency did not affect NAFLD‐independent liver injury and fibrosis in MDR2 KO or CCl4‐treated mice. Finally, we treated wild‐type mice exposed to NASH‐inducing diet with an oral MPO inhibitor. Pharmacological MPO inhibition not only reduced markers of MPO‐mediated liver damage, serum alanine aminotransferase levels, and hepatic steatosis, but also significantly decreased NASH‐induced liver fibrosis. MPO inhibitor treatment, but not MPO deficiency, significantly altered gut microbiota including a significant expansion of Akkermansia muciniphila. Conclusions: MPO specifically promotes NASH‐induced liver fibrosis. Pharmacological MPO inhibition attenuates NASH progression and NASH‐induced liver fibrosis in mice and is associated with beneficial changes of intestinal microbiota.
url https://doi.org/10.1002/hep4.1566
work_keys_str_mv AT anjachristinakoop therapeutictargetingofmyeloperoxidaseattenuatesnashinmice
AT ninadoreenthiele therapeutictargetingofmyeloperoxidaseattenuatesnashinmice
AT davidsteins therapeutictargetingofmyeloperoxidaseattenuatesnashinmice
AT erikmichaelsson therapeutictargetingofmyeloperoxidaseattenuatesnashinmice
AT maltewehmeyer therapeutictargetingofmyeloperoxidaseattenuatesnashinmice
AT ludgerscheja therapeutictargetingofmyeloperoxidaseattenuatesnashinmice
AT babettsteglich therapeutictargetingofmyeloperoxidaseattenuatesnashinmice
AT samuelhuber therapeutictargetingofmyeloperoxidaseattenuatesnashinmice
AT julianschulzezurwiesch therapeutictargetingofmyeloperoxidaseattenuatesnashinmice
AT ansgarwlohse therapeutictargetingofmyeloperoxidaseattenuatesnashinmice
AT jorgheeren therapeutictargetingofmyeloperoxidaseattenuatesnashinmice
AT johanneskluwe therapeutictargetingofmyeloperoxidaseattenuatesnashinmice
_version_ 1724253088589545472