File Download

There are no files associated with this item.

  Links for fulltext
     (May Require Subscription)
Supplementary

Article: Neutrophil Extracellular Traps Mediate Acute Liver Failure in Regulation of miR-223/Neutrophil Elastase Signaling in Mice

TitleNeutrophil Extracellular Traps Mediate Acute Liver Failure in Regulation of miR-223/Neutrophil Elastase Signaling in Mice
Authors
KeywordsHepatocellular Injury
NET Inhibitors
Neutrophil Degranulation
Issue Date1-Jun-2022
PublisherElsevier
Citation
Cellular and Molecular Gastroenterology and Hepatology, 2022, v. 14, n. 3, p. 587-607 How to Cite?
Abstract

Background & Aims

Marked enhancement of neutrophil infiltration in the liver is a hallmark of acute liver failure (ALF), a severe life-threatening disease with varying etiologies. However, the mechanisms and pathophysiological role corresponding to hepatic neutrophil infiltration during ALF development remain poorly characterized.

Methods

Experimental ALF was induced in 10-week-old male microRNA-223 (miR-223) knockout (KO) mice, neutrophil elastase (NE) KO mice, and wild-type controls by intraperitoneal injection of galactosamine hydrochloride and lipopolysaccharide. Age-matched mice were injected with phosphate-buffered saline and served as vehicle controls.

Results

Mouse liver with ALF showed evident formation of neutrophil extracellular traps (NETs), which were enhanced markedly in miR-223 KO mice. The blockade of NETs by pharmacologic inhibitor GSK484 significantly attenuated neutrophil infiltration and massive necrosis in mouse liver with ALF. ALF-related hepatocellular damage and mortality in miR-223 KO mice were aggravated significantly and accompanied by potentiated neutrophil infiltration in the liver when compared with wild-type controls. Transcriptomic analyses showed that miR-223 deficiency in bone marrow predominantly caused the enrichment of pathways involved in neutrophil degranulation. Likewise, ALF-induced hepatic NE enrichment was potentiated in miR-223 KO mice. Genetic ablation of NE blunted the formation of NETs in parallel with significant attenuation of ALF in mice. Pharmaceutically, pretreatment with the NE inhibitor sivelestat protected mice against ALF.

Conclusions

The present study showed the miR-223/NE axis as a key modulator of NETs, thereby exacerbating oxidative stress and neutrophilic inflammation to potentiate hepatocellular damage and liver necrosis in ALF development, and offering potential targets against ALF.


Persistent Identifierhttp://hdl.handle.net/10722/338263
ISSN
2021 Impact Factor: 8.797
2020 SCImago Journal Rankings: 3.417
ISI Accession Number ID

 

DC FieldValueLanguage
dc.contributor.authorYe, DW-
dc.contributor.authorYao, JY-
dc.contributor.authorDu WF,-
dc.contributor.authorChen, CS-
dc.contributor.authorYang, Y-
dc.contributor.authorYan, KX-
dc.contributor.authorLi, JF-
dc.contributor.authorXu, Y-
dc.contributor.authorZang, SF-
dc.contributor.authorZhang, YY-
dc.contributor.authorRong, XL-
dc.contributor.authorZhang, RX-
dc.contributor.authorXu, AM-
dc.contributor.authorGuo, J-
dc.date.accessioned2024-03-11T10:27:32Z-
dc.date.available2024-03-11T10:27:32Z-
dc.date.issued2022-06-01-
dc.identifier.citationCellular and Molecular Gastroenterology and Hepatology, 2022, v. 14, n. 3, p. 587-607-
dc.identifier.issn2352-345X-
dc.identifier.urihttp://hdl.handle.net/10722/338263-
dc.description.abstract<h3>Background & Aims</h3><p>Marked enhancement of neutrophil infiltration in the liver is a hallmark of acute liver failure (ALF), a severe life-threatening disease with varying etiologies. However, the mechanisms and pathophysiological role corresponding to hepatic neutrophil infiltration during ALF development remain poorly characterized.</p><h3>Methods</h3><p>Experimental ALF was induced in 10-week-old male microRNA-223 (miR-223) knockout (KO) mice, neutrophil elastase (NE) KO mice, and wild-type controls by intraperitoneal injection of galactosamine hydrochloride and lipopolysaccharide. Age-matched mice were injected with phosphate-buffered saline and served as vehicle controls.</p><h3>Results</h3><p>Mouse liver with ALF showed evident formation of neutrophil extracellular traps (NETs), which were enhanced markedly in miR-223 KO mice. The blockade of NETs by pharmacologic inhibitor GSK484 significantly attenuated neutrophil infiltration and massive necrosis in mouse liver with ALF. ALF-related hepatocellular damage and mortality in miR-223 KO mice were aggravated significantly and accompanied by potentiated neutrophil infiltration in the liver when compared with wild-type controls. Transcriptomic analyses showed that miR-223 deficiency in bone marrow predominantly caused the enrichment of pathways involved in neutrophil degranulation. Likewise, ALF-induced hepatic NE enrichment was potentiated in miR-223 KO mice. Genetic ablation of NE blunted the formation of NETs in parallel with significant attenuation of ALF in mice. Pharmaceutically, pretreatment with the NE inhibitor sivelestat protected mice against ALF.</p><h3>Conclusions</h3><p>The present study showed the miR-223/NE axis as a key modulator of NETs, thereby exacerbating oxidative stress and neutrophilic inflammation to potentiate hepatocellular damage and liver necrosis in ALF development, and offering potential targets against ALF.</p>-
dc.languageeng-
dc.publisherElsevier-
dc.relation.ispartofCellular and Molecular Gastroenterology and Hepatology-
dc.rightsThis work is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License.-
dc.subjectHepatocellular Injury-
dc.subjectNET Inhibitors-
dc.subjectNeutrophil Degranulation-
dc.titleNeutrophil Extracellular Traps Mediate Acute Liver Failure in Regulation of miR-223/Neutrophil Elastase Signaling in Mice-
dc.typeArticle-
dc.identifier.doi10.1016/j.jcmgh.2022.05.012-
dc.identifier.pmid35660025-
dc.identifier.scopuseid_2-s2.0-85134665365-
dc.identifier.volume14-
dc.identifier.issue3-
dc.identifier.spage587-
dc.identifier.epage607-
dc.identifier.eissn2352-345X-
dc.identifier.isiWOS:000860358300005-
dc.publisher.placeSAN DIEGO-
dc.identifier.issnl2352-345X-

Export via OAI-PMH Interface in XML Formats


OR


Export to Other Non-XML Formats