Shear stress associated with cardiopulmonary bypass induces expression of inflammatory cytokines and necroptosis in monocytes

JCI Insight. 2021 Jan 11;6(1):e141341. doi: 10.1172/jci.insight.141341.

Abstract

Cardiopulmonary bypass (CPB) is required during most cardiac surgeries. CBP drives systemic inflammation and multiorgan dysfunction that is especially severe in neonatal patients. Limited understanding of molecular mechanisms underlying CPB-associated inflammation presents a significant barrier to improve clinical outcomes. To better understand these clinical issues, we performed mRNA sequencing on total circulating leukocytes from neonatal patients undergoing CPB. Our data identify myeloid cells, particularly monocytes, as the major cell type driving transcriptional responses to CPB. Furthermore, IL-8 and TNF-α were inflammatory cytokines robustly upregulated in leukocytes from both patients and piglets exposed to CPB. To delineate the molecular mechanism, we exposed THP-1 human monocytic cells to CPB-like conditions, including artificial surfaces, high shear stress, and cooling/rewarming. Shear stress was found to drive cytokine upregulation via calcium-dependent signaling pathways. We also observed that a subpopulation of THP-1 cells died via TNF-α-mediated necroptosis, which we hypothesize contributes to post-CPB inflammation. Our study identifies a shear stress-modulated molecular mechanism that drives systemic inflammation in pediatric CPB patients. These are also the first data to our knowledge to demonstrate that shear stress causes necroptosis. Finally, we observe that calcium and TNF-α signaling are potentially novel targets to ameliorate post-CPB inflammation.

Keywords: Calcium signaling; Cardiology; Cytokines; Inflammation; Surgery.

Publication types

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

MeSH terms

  • Animals
  • Animals, Newborn
  • Calcium Signaling
  • Cardiopulmonary Bypass / adverse effects*
  • Cytokines / biosynthesis
  • Cytokines / genetics*
  • Female
  • Heart Defects, Congenital / surgery
  • Humans
  • Infant
  • Infant, Newborn
  • Inflammation Mediators / metabolism
  • Interleukin-8 / biosynthesis
  • Interleukin-8 / genetics
  • Male
  • Models, Animal
  • Monocytes / immunology*
  • Monocytes / pathology*
  • Monocytes / physiology
  • Necroptosis / genetics
  • Necroptosis / physiology
  • RNA-Seq
  • Stress, Mechanical
  • Sus scrofa
  • Systemic Inflammatory Response Syndrome / etiology
  • Systemic Inflammatory Response Syndrome / genetics
  • Systemic Inflammatory Response Syndrome / immunology
  • THP-1 Cells
  • Tumor Necrosis Factor-alpha / biosynthesis
  • Tumor Necrosis Factor-alpha / genetics
  • Up-Regulation

Substances

  • Cytokines
  • Inflammation Mediators
  • Interleukin-8
  • Tumor Necrosis Factor-alpha