4.3 Article

Trimethylamine-N-oxide-stimulated hepatocyte-derived exosomes promote inflammation and endothelial dysfunction through nuclear factor-kappa B signaling

Journal

ANNALS OF TRANSLATIONAL MEDICINE
Volume 9, Issue 22, Pages -

Publisher

AME PUBLISHING COMPANY
DOI: 10.21037/atm-21-5043

Keywords

Trimethylamine-N-oxide; hepatocyte-derived exosomes; inflammation; endothelial function; microRNA

Funding

  1. National Key Research and Development Program of China [2018YFC1002600, 2020YFC2008005]
  2. Guangdong peak project [DFJH2019]
  3. Science and Technology Projects in Guangzhou [202102021149, 202002020030]
  4. Postdoctoral Scientific Research Start-up Fund Project of Guangdong Provincial People's Hospital [BY012021052]
  5. National Nature Science Foundation of China [82100451]

Ask authors/readers for more resources

Research shows that TMAO can stimulate hepatocytes to release Exos that induce inflammation and cell apoptosis, impair cell migration, and inhibit endothelium-dependent vasodilation in VECs. The miRNAs carried by these TMAO-Exos are different from those in the TMAO-free group, potentially targeting genes involved in inflammation and endothelial function. Activation of the NF-Kappa B signaling pathway by TMAO-Exos is also observed.
Background: Trimethylamine-N-oxide (TMAO) has been proven to be a new proatherogenic compound for promoting inflammation and endothelial dysfunction. Hepatocyte-derived exosomes (Exos), including those derived from hepatocytes, play a pivotal role in the regulation of inflammation and endothelial function. As TMAO is produced in the liver, hepatocytes may be the potential target of TMAO. However, it is not yet clear whether TMAO can directly stimulate hepatocytes to produce Exos to mediate the detrimental effects of TMAO on vascular endothelial cells (VECs). Methods: Hepatocytes treated with TMAO and Exos (TMAO-Exos) were isolated from the supernatant, and added to human aortic endothelial cells (HAECs). The expressions of interleukin-6 (IL-6), monocyte chemotactic protein-1 (MCP-1), and tumor necrosis factor-alpha (TNF-alpha) were detected by quantitative polymerase chain reaction (qPCR). Cell apoptosis was evaluated using Hoechst 33342 staining and flow cytometry assay, and cell migration was assessed by scratch and transwell assay. C57BL/6 mice were treated with Exos for 24 h and the thoracic aortas were isolated, then the in vitro aortic ring bioassay was conducted to determine the changes of vasodilation. The expressions of cluster of differentiation 81, tumor susceptibility gene 101, nuclear factor-kappa B (NF-Kappa B) p65, and Phospho-NF-Kappa B p65 were detected by western blotting. The micro ribonucleic acid (miRNA) profiles of the Exos were then identified using RNA-sequencing and validated by qPCR. The miRNA-messenger RNA networks were constructed, and the biological functions of the target genes were annotated using bioinformatics methods. Results: TMAO was found to stimulate hepatocytes to release Exos that could be taken up by HAECs, thus inducing inflammation and cell apoptosis, impairing cell migration, and inhibiting endothelium-dependent vasodilation. Additionally, the miRNAs such as miR-302d-3p carried by the TMAO-Exos were quite different to those in the TMAO-free group. A further analysis showed that the potential target genes for these miRNAs, such as mitogen-activated protein kinase 8, caspase 9 and BCL2-like 11, appeared to be involved with inflammation and endothelial function. Finally, we found that NF-Kappa B signaling could be activated by TMAO-Exos. Conclusions: These novel findings provide evidence that TMAO can indirectly talk to VECs by promoting hepatocytes to produce Exos that carry important genetic information.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.3
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available