1. Academic Validation
  2. PFKFB3 promotes endotoxemia-induced myocardial dysfunction through inflammatory signaling and apoptotic induction

PFKFB3 promotes endotoxemia-induced myocardial dysfunction through inflammatory signaling and apoptotic induction

  • Toxicol Appl Pharmacol. 2019 Apr 1;368:26-36. doi: 10.1016/j.taap.2019.02.007.
Wen Tian 1 Hong-Sheng Guo 1 Chong-Yao Li 2 Wei Cao 3 Xue-Ying Wang 1 Dan Mo 1 Xiao-Wei Hao 1 Ying-Da Feng 1 Yang Sun 1 Fan Lei 2 Hui-Nan Zhang 1 Ming-Gao Zhao 1 Xiao-Qiang Li 4
Affiliations

Affiliations

  • 1 Department of Pharmacology and Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, School of Pharmacy, Fourth Military Medical University, Xi'an, China.
  • 2 School of Basic Medical Sciences, Hubei University of Science and Technology, Xianning, China.
  • 3 Shaanxi Key Laboratory of Natural Products & Chemical Biology, School of Chemistry & Pharmacy, Northwest A&F University, Yangling, China. Electronic address: [email protected].
  • 4 Department of Pharmacology and Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, School of Pharmacy, Fourth Military Medical University, Xi'an, China. Electronic address: [email protected].
Abstract

Cardiac dysfunction is a vital complication during endotoxemia (ETM). Accumulating evidence suggests that enhanced glycolytic metabolism promotes inflammatory and myocardial diseases. In this study, we performed deep mRNA sequencing analysis on the hearts of control and lipopolysaccharide (LPS)-challenged mice (40 mg/kg, i.p.) and identified that the glycolytic Enzyme, 6-phosphofructo-2-kinase (PFK-2)/fructose-2,6-bisphosphatase 3 (PFKFB3) might play an indispensable role in ETM-induced cardiac damage. Quantitative Real-Time PCR validated the transcriptional upregulation of PFKFB3 in the myocardium of LPS-challenged mice and immunoblotting and immunostaining assays confirmed that LPS stimulation markedly increased the expression of PFKFB3 at the protein level both in vivo and in vitro. The potent antagonist 3-(3pyridinyl)-1-(4-pyridinyl)-2-propen-1-one (3PO) was used to block PFKFB3 activity in vivo (50 mg/kg, i.p.) and in vitro (10 μM). Echocardiographic analysis and TUNEL staining showed that 3PO significantly alleviated LPS-induced cardiac dysfunction and apoptotic injury in vivo. 3PO also suppressed the LPS-induced secretion of tumor necrosis factor-α, interleukin (IL)-1β, IL-6 and lactate in the serum, in addition to lactate in the myocardium. PFKFB3 inhibition also diminished the nuclear translocation and phosphorylation of transcription factor nuclear factor-κB (NF-κB) in both adult cardiomyocytes and HL-1 cells. Furthermore, immunoblotting analysis showed that 3PO inhibited LPS-induced apoptotic induction in cardiomyocytes. Taken together, these findings demonstrate that PFKFB3 participates in LPS-induced cardiac dysfunction via mediating inflammatory and apoptotic signaling pathway.

Keywords

3-(3pyridinyl)-1-(4-pyridinyl)-2-propen-1-one (3PO); Apoptosis; Cardiac dysfunction; Inflammation; PFKFB3.

Figures
Products
  • Cat. No.
    Product Name
    Description
    Target
    Research Area
  • HY-19824
    99.56%, PFKFB3 Blocker