Krüppel-like factor 14 deletion in myeloid cells accelerates atherosclerotic lesion development

Cardiovasc Res. 2022 Jan 29;118(2):475-488. doi: 10.1093/cvr/cvab027.

Abstract

Aims: Atherosclerosis is the dominant pathologic basis of many cardiovascular diseases. Large genome-wide association studies have identified that single-nucleotide polymorphisms proximal to Krüppel-like factor 14 (KLF14), a member of the zinc finger family of transcription factors, are associated with higher cardiovascular risks. Macrophage dysfunction contributes to atherosclerosis development and has been recognized as a potential therapeutic target for treating many cardiovascular diseases. Herein, we address the biologic function of KLF14 in macrophages and its role during the development of atherosclerosis.

Methods and results: KLF14 expression was markedly decreased in cholesterol loaded foam cells, and overexpression of KLF14 significantly increased cholesterol efflux and inhibited the inflammatory response in macrophages. We generated myeloid cell-selective Klf14 knockout (Klf14LysM) mice in the ApoE-/- background for the atherosclerosis study. Klf14LysMApoE-/- and litter-mate control mice (Klf14fl/flApoE-/-) were placed on the Western Diet for 12 weeks to induce atherosclerosis. Macrophage Klf14 deficiency resulted in increased atherosclerosis development without affecting the plasma lipid profiles. Klf14-deficient peritoneal macrophages showed significantly reduced cholesterol efflux resulting in increased lipid accumulation and exacerbated inflammatory response. Mechanistically, KLF14 upregulates the expression of a key cholesterol efflux transporter, ABCA1 (ATP-binding cassette transporter A1), while it suppresses the expression of several critical components of the inflammatory cascade. In macrophages, activation of KLF14 by its activator, perhexiline, a drug clinically used to treat angina, significantly inhibited the inflammatory response and increased cholesterol efflux in a KLF14-dependent manner in macrophages without triggering hepatic lipogenesis.

Conclusions: This study provides insights into the anti-atherosclerotic effects of myeloid KLF14 through promoting cholesterol efflux and suppressing the inflammatory response. Activation of KLF14 may represent a potential new therapeutic approach to prevent or treat atherosclerosis.

Keywords: Krüppel-like factor • Atherosclerosis• Cholesterol efflux • Inflammation.

Publication types

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

MeSH terms

  • ATP Binding Cassette Transporter 1 / metabolism
  • Animals
  • Aorta / immunology
  • Aorta / metabolism*
  • Aorta / pathology
  • Aortic Diseases / genetics
  • Aortic Diseases / immunology
  • Aortic Diseases / metabolism*
  • Aortic Diseases / pathology
  • Atherosclerosis / genetics
  • Atherosclerosis / immunology
  • Atherosclerosis / metabolism*
  • Atherosclerosis / pathology
  • Cholesterol / metabolism
  • Disease Models, Animal
  • Disease Progression
  • Female
  • Hep G2 Cells
  • Humans
  • Interleukin-1beta / metabolism
  • Kruppel-Like Transcription Factors / deficiency*
  • Kruppel-Like Transcription Factors / genetics
  • Macrophages / immunology
  • Macrophages / metabolism*
  • Macrophages / pathology
  • Male
  • Mice
  • Mice, Inbred C57BL
  • Mice, Knockout, ApoE
  • Plaque, Atherosclerotic*
  • Signal Transduction
  • THP-1 Cells
  • Transcription Factor RelA / metabolism

Substances

  • ABCA1 protein, human
  • ABCA1 protein, mouse
  • ATP Binding Cassette Transporter 1
  • IL1B protein, human
  • IL1B protein, mouse
  • Interleukin-1beta
  • KLF14 protein, human
  • Klf14 protein, mouse
  • Kruppel-Like Transcription Factors
  • RELA protein, human
  • Rela protein, mouse
  • Transcription Factor RelA
  • Cholesterol