Mitochondria-targeted drugs enhance Nlrp3 inflammasome-dependent IL-1β secretion in association with alterations in cellular redox and energy status

Free Radic Biol Med. 2013 Jul:60:233-45. doi: 10.1016/j.freeradbiomed.2013.01.025. Epub 2013 Jan 29.

Abstract

The Nlrp3 inflammasome is activated in response to an array of environmental and endogenous molecules leading to caspase-1-dependent IL-1β processing and secretion by myeloid cells. Several identified Nlrp3 inflammasome activators also trigger reactive oxygen species (ROS) production. However, the initial concept that NADPH oxidases are the primary source of ROS production during inflammasome activation is becoming less accepted. Therefore, the importance of mitochondria-derived ROS has been recently explored. In this study, we explore the impact of mitochondria dysfunction and ROS production on Nlrp3 inflammasome stimulation and IL-1β secretion induced by serum amyloid A (SAA) in primary mouse peritoneal macrophages. To induce mitochondrial dysfunction, we utilized antimycin A, which blocks electron flow at complex III, and carbonyl cyanide-p-trifluoromethoxyphenylhydrazone (FCCP), a mitochondrial oxidative phosphorylation uncoupler. We also utilized a superoxide dismutase mimetic, MnTBAP, which targets the mitochondria, as well as the broad-spectrum antioxidants DPI (diphenyleneiodonium chloride) and ebselen. Our findings demonstrate that SAA alone induces mitochondrial ROS in a time-dependent manner. We observed that MnTBAP and ebselen blocked IL-1β secretion caused by SAA only when added before stimulation, and DPI augmented IL-1β secretion. Surprisingly, these effects were not directly related to intracellular or mitochondrial ROS levels. We also found that mitochondria-targeted drugs increased IL-1β secretion regardless of their impact on mitochondrial function and ROS levels, suggesting that mitochondrial ROS-dependent and -independent mechanisms play a role in the Nlrp3 inflammasome/IL-1β secretion axis in SAA-stimulated cells. Finally, we found that FCCP significantly sustained the association of the Nlrp3 inflammasome complex, which could explain the most robust effect among the drugs tested in enhancing IL-1β secretion in SAA-treated cells. Overall, our data suggest that the Nlrp3 inflammasome/IL-1β secretion axis is a very highly regulated inflammatory pathway that is susceptible not only to changes in mitochondrial or intracellular ROS, but also to changes in overall mitochondrial function.

MeSH terms

  • Animals
  • Carbonyl Cyanide p-Trifluoromethoxyphenylhydrazone / administration & dosage
  • Carrier Proteins / genetics
  • Carrier Proteins / metabolism*
  • Energy Metabolism / drug effects
  • Inflammasomes / drug effects
  • Inflammasomes / genetics
  • Inflammasomes / metabolism*
  • Interleukin-1beta / genetics
  • Interleukin-1beta / metabolism*
  • Macrophages, Peritoneal / cytology
  • Macrophages, Peritoneal / drug effects
  • Mice
  • Mitochondria / drug effects
  • Mitochondria / metabolism*
  • Mitochondria / pathology
  • NLR Family, Pyrin Domain-Containing 3 Protein
  • Oxidation-Reduction / drug effects
  • Primary Cell Culture
  • Reactive Oxygen Species / metabolism
  • Serum Amyloid A Protein / pharmacology

Substances

  • Carrier Proteins
  • Inflammasomes
  • Interleukin-1beta
  • NLR Family, Pyrin Domain-Containing 3 Protein
  • Nlrp3 protein, mouse
  • Reactive Oxygen Species
  • Serum Amyloid A Protein
  • Carbonyl Cyanide p-Trifluoromethoxyphenylhydrazone