Chemical hypoxia induces pro-inflammatory signals in fat-laden hepatocytes and contributes to cellular crosstalk with Kupffer cells through extracellular vesicles

Biochim Biophys Acta Mol Basis Dis. 2020 Jun 1;1866(6):165753. doi: 10.1016/j.bbadis.2020.165753. Epub 2020 Feb 29.

Abstract

Background: Obstructive sleep apnea syndrome (OSAS) is associated to intermittent hypoxia (IH) and is an aggravating factor of non-alcoholic fatty liver disease (NAFLD). We investigated the effects of hypoxia in both in vitro and in vivo models of NAFLD.

Methods: Primary rat hepatocytes treated with free fatty acids (FFA) were subjected to chemically induced hypoxia (CH) using the hypoxia-inducible factor-1 alpha (HIF-1α) stabilizer cobalt chloride (CoCl2). Triglyceride (TG) content, mitochondrial superoxide production, cell death rates, cytokine and inflammasome components gene expression and protein levels of cleaved caspase-1 were assessed. Also, Kupffer cells (KC) were treated with conditioned medium (CM) and extracellular vehicles (EVs) from hypoxic fat-laden hepatic cells. The choline deficient L-amino acid defined (CDAA)-feeding model used to assess the effects of IH on experimental NAFLD in vivo.

Results: Hypoxia induced HIF-1α in cells and animals. Hepatocytes exposed to FFA and CoCl2 exhibited increased TG content and higher cell death rates as well as increased mitochondrial superoxide production and mRNA levels of pro-inflammatory cytokines and of inflammasome-components interleukin-1β, NLRP3 and ASC. Protein levels of cleaved caspase-1 increased in CH-exposed hepatocytes. CM and EVs from hypoxic fat-laden hepatic cells evoked a pro-inflammatory phenotype in KC. Livers from CDAA-fed mice exposed to IH exhibited increased mRNA levels of pro-inflammatory and inflammasome genes and increased levels of cleaved caspase-1.

Conclusion: Hypoxia promotes inflammatory signals including inflammasome/caspase-1 activation in fat-laden hepatocytes and contributes to cellular crosstalk with KC by release of EVs. These mechanisms may underlie the aggravating effect of OSAS on NAFLD. [Abstract word count: 257].

Keywords: Fatty liver disease; Hepatology; Hypoxia, liver injury, steatosis, apoptosis; Nonalcoholic fatty liver disease.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Caspase 1 / genetics
  • Choline Deficiency / genetics
  • Choline Deficiency / metabolism
  • Choline Deficiency / pathology
  • Cobalt / toxicity
  • Disease Models, Animal
  • Extracellular Vesicles / genetics
  • Extracellular Vesicles / metabolism
  • Extracellular Vesicles / pathology
  • Fatty Acids, Nonesterified / pharmacology
  • Gene Expression Regulation / drug effects
  • Hepatocytes / metabolism
  • Hepatocytes / pathology
  • Humans
  • Hypoxia / chemically induced
  • Hypoxia / genetics*
  • Hypoxia / metabolism
  • Hypoxia / pathology
  • Hypoxia-Inducible Factor 1, alpha Subunit / genetics*
  • Inflammasomes / genetics
  • Inflammation / chemically induced
  • Inflammation / genetics
  • Inflammation / metabolism
  • Inflammation / pathology
  • Interleukin-1beta / genetics
  • Kupffer Cells / metabolism
  • Kupffer Cells / pathology
  • Mice
  • NLR Family, Pyrin Domain-Containing 3 Protein / genetics*
  • Non-alcoholic Fatty Liver Disease / complications
  • Non-alcoholic Fatty Liver Disease / genetics*
  • Non-alcoholic Fatty Liver Disease / metabolism
  • Non-alcoholic Fatty Liver Disease / pathology
  • Rats
  • Sleep Apnea, Obstructive / etiology
  • Sleep Apnea, Obstructive / genetics*
  • Sleep Apnea, Obstructive / metabolism
  • Sleep Apnea, Obstructive / pathology
  • Triglycerides / genetics

Substances

  • Fatty Acids, Nonesterified
  • Hif1a protein, rat
  • Hypoxia-Inducible Factor 1, alpha Subunit
  • IL1B protein, rat
  • Inflammasomes
  • Interleukin-1beta
  • NLR Family, Pyrin Domain-Containing 3 Protein
  • Nlrp3 protein, rat
  • Triglycerides
  • Cobalt
  • Caspase 1
  • cobaltous chloride