Abstract

Growing clinical and experimental evidence suggests that sterile inflammation contributes to alcoholic liver disease (ALD). High mobility group box-1 (HMGB1) is highly induced during liver injury; however, a link between this alarmin and ALD has not been established. Thus, the aim of this work was to determine whether HMGB1 contributes to the pathogenesis of ALD. Liver biopsies from patients with ALD showed a robust increase in HMGB1 expression and translocation, which correlated with disease stage, compared with healthy explants. Similar findings were observed in chronic ethanol-fed wild-type (WT) mice. Using primary cell culture, we validated the ability of hepatocytes from ethanol-fed mice to secrete a large amount of HMGB1. Secretion was time- and dose-dependent and responsive to prooxidants and antioxidants. Selective ablation of Hmgb1 in hepatocytes protected mice from alcohol-induced liver injury due to increased carnitine palmitoyltransferase-1, phosphorylated 5'AMP-activated protein kinase-α, and phosphorylated peroxisome proliferator-activated receptor-α expression along with elevated LDL plus VLDL export. Native and post-translationally modified HMGB1 were detected in humans and mice with ALD. In liver and serum from control mice and in serum from healthy volunteers, the lysine residues within the peptides containing nuclear localization signals (NLSs) 1 and 2 were non-acetylated, and all cysteine residues were reduced. However, in livers from ethanol-fed mice, in addition to all thiol/non-acetylated isoforms of HMGB1, we observed acetylated NLS1 and NLS2, a unique phosphorylation site in serine 35, and an increase in oxidation of HMGB1 to the disulfide isoform. In serum from ethanol-fed mice and from patients with ALD, there was disulfide-bonded hyperacetylated HMGB1, disulfide-bonded non-acetylated HMGB1, and HMGB1 phosphorylated in serine 35. Hepatocytes appeared to be a major source of these HMGB1 isoforms. Thus, hepatocyte HMGB1 participates in the pathogenesis of ALD and undergoes post-translational modifications (PTMs) that could condition its toxic effects.

Highlights

  • High mobility group box-1 (HMGB1) is a proinflammatory cytokine produced in response to tissue injury, but its role in alcoholic liver disease (ALD) is unknown

  • Because inflammation is a key component contributing to the pathogenesis of ALD and HMGB1 is an early proinflammatory cytokine up-regulated in response to sterile and non-sterile tissue injury, the aims of this study were to determine whether HMGB1 increases in ALD, quantify the extent of HMGB1 translocation from the nucleus to the cytoplasm in ALD, dissect whether HMGB1 induction in hepatocytes is essential for alcohol-induced liver injury, dissect the mechanism involved, identify the post-translational modifications (PTMs) of HMGB1 in ALD, and recognize whether native HMGB1 and post-translationally modified HMGB1 are secreted into the extracellular space under alcohol consumption

  • Liver Biopsies from Patients with Acute Alcoholic Steatohepatitis (ASH) Superimposed on ALD and Cirrhosis Show Significant and Progressive Increase in HMGB1 Expression and Translocation from the Nucleus to the Cytoplasm Compared with Wedge Samples from Healthy Human Lobectomy Specimens— To determine whether HMGB1 increases in ALD, wedge samples from healthy human lobectomy specimens and liver needle biopsies from patients with clinically proven acute ASH superimposed on ALD and cirrhosis, as shown by Hematoxylin and eosin (H&E) and Sirius red/fast green staining (Fig. 1A), were analyzed for HMGB1 expression

Read more

Summary

Introduction

HMGB1 is a proinflammatory cytokine produced in response to tissue injury, but its role in ALD is unknown. Results: HMGB1 increases; translocates; and undergoes acetylation, phosphorylation, and oxidation in ALD. HMGB1 ablation in hepatocytes protects against steatosis and injury in ALD. Significance: Dissecting how the increase in HMGB1 causes hepatotoxicity is key for understanding the pathogenesis of ALD. Liver biopsies from patients with ALD showed a robust increase in HMGB1 expression and translocation, which correlated with disease stage, compared with healthy explants. We validated the ability of hepatocytes from ethanol-fed mice to secrete a large amount of HMGB1. Selective ablation of Hmgb in hepatocytes protected mice from alcohol-induced liver injury due to increased carnitine palmitoyltransferase-1, phosphorylated 5؅AMP-activated protein kinase-␣, and phosphorylated peroxisome proliferator-activated receptor-␣ expression along with elevated LDL plus VLDL export.

Objectives
Results
Conclusion
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.