Pressure overload induces IL-18 and IL-18R expression, but markedly suppresses IL-18BP expression in a rabbit model. IL-18 potentiates TNF-α-induced cardiomyocyte death

J Mol Cell Cardiol. 2014 Oct:75:141-51. doi: 10.1016/j.yjmcc.2014.07.007. Epub 2014 Aug 7.

Abstract

Recurrent or sustained inflammation plays a causal role in the development and progression of left ventricular hypertrophy (LVH) and its transition to failure. Interleukin (IL)-18 is a potent pro-hypertrophic inflammatory cytokine. We report that induction of pressure overload in the rabbit, by constriction of the descending thoracic aorta induces compensatory hypertrophy at 4weeks (mass/volume ratio: 1.7±0.11) and ventricular dilatation indicative of heart failure at 6weeks (mass/volume ratio: 0.7±0.04). In concordance with this, fractional shortening was preserved at 4weeks, but markedly attenuated at 6weeks. We cloned rabbit IL-18, IL-18Rα, IL-18Rβ, and IL-18 binding protein (IL-18BP) cDNA, and show that pressure overload, while enhancing IL-18 and IL-18R expression in hypertrophied and failing hearts, markedly attenuated the level of expression of the endogenous IL-18 antagonist IL-18BP. Cyclical mechanical stretch (10% cyclic equibiaxial stretch, 1Hz) induced hypertrophy of primary rabbit cardiomyocytes in vitro and enhanced ANP, IL-18, and IL-18Rα expression. Further, treatment with rhIL-18 induced its own expression and that of IL-18Rα via AP-1 activation, and induced cardiomyocyte hypertrophy in part via PI3K/Akt/GATA4 signaling. In contrast, IL-18 potentiated TNF-α-induced cardiomyocyte death, and by itself induced cardiac endothelial cell death. These results demonstrate that pressure overload is associated with enhanced IL-18 and its receptor expression in hypertrophied and failingrabbit hearts. Since IL-18BP expression is markedly inhibited, our results indicate a positive amplification in IL-18 proinflammatory signaling during pressure overload, and suggest IL-18 as a potential therapeutic target in pathological hypertrophy and cardiac failure.

Keywords: Cardiac failure; Cyclical stretch; Inflammation; Interleukins; Myocardial hypertrophy.

Publication types

  • Research Support, N.I.H., Extramural
  • Research Support, U.S. Gov't, Non-P.H.S.

MeSH terms

  • Animals
  • Cardiomegaly / diagnostic imaging
  • Cardiomegaly / pathology
  • Cell Death / drug effects
  • Disease Models, Animal
  • Endothelial Cells / drug effects
  • Endothelial Cells / pathology
  • Heart Failure / metabolism
  • Humans
  • Intercellular Signaling Peptides and Proteins / genetics
  • Intercellular Signaling Peptides and Proteins / metabolism*
  • Interleukin-18 / genetics
  • Interleukin-18 / metabolism*
  • Male
  • Molecular Sequence Data
  • Myocytes, Cardiac / drug effects
  • Myocytes, Cardiac / metabolism
  • Myocytes, Cardiac / pathology*
  • Pressure
  • RNA, Messenger / genetics
  • RNA, Messenger / metabolism
  • Rabbits
  • Receptors, Interleukin-18 / genetics
  • Receptors, Interleukin-18 / metabolism*
  • Stress, Mechanical
  • Tumor Necrosis Factor-alpha / pharmacology*
  • Ultrasonography

Substances

  • Intercellular Signaling Peptides and Proteins
  • Interleukin-18
  • RNA, Messenger
  • Receptors, Interleukin-18
  • Tumor Necrosis Factor-alpha
  • interleukin-18 binding protein

Associated data

  • GENBANK/JF271681
  • GENBANK/JF271682
  • GENBANK/JF271683
  • GENBANK/JQ657816
  • GENBANK/JQ657817