4.7 Article

Preparation of a miR-155-activating nucleic acid nanoflower to study the molecular mechanism of miR-155 in inflammation

Journal

MOLECULAR MEDICINE
Volume 28, Issue 1, Pages -

Publisher

SPRINGER
DOI: 10.1186/s10020-022-00495-4

Keywords

Nucleic acid nanoflowers; miR-155; RNA activation; Inflammation

Funding

  1. Project of Science and Technology of Fujian Province of China [2019J05094, 2020NZ010008]
  2. basic operating expenses of central universities-Huaqiao University Young and Middle-aged Teachers Science and Technology Innovation Funding Program [ZQN-PY319]

Ask authors/readers for more resources

The molecular mechanisms underlying inflammation remain unclear. This study designed a nucleic acid nanoflowers (NFs) complex to directly activate inflammatory genes and investigate the molecular mechanism of inflammation. The findings suggest that miR-155 and its target genes act as a molecular switch role in the development of inflammation.
At present, the molecular mechanisms underlying inflammation remain unclear. In recent years, research on inflammation has focused on stimulating cell inflammation by using exogenous pro-inflammatory substances such as lipopolysaccharide (LPS) or inflammatory factors. To investigate the molecular mechanism of inflammation from a new perspective, we designed a nucleic acid nanoflowers (NFs) complex to directly activate inflammatory genes to study the inflammatory response without the need for external microbial factors to trigger an inflammatory response. An RNAa-type target gene-activated NFs was designed. Human umbilical vein endothelial cells (HUVECs) were transfected with NFs carrying small activating RNA (saRNAs) to directly co-activate microRNA (miR)-155 and SHIP1 genes. After RNA activation (RNAa)-type NFs were transferred into HUVECs, the expression of miR-155 and pro-inflammatory and cancer-related factors increased, anti-inflammatory factors were reduced, cell proliferation increased, and cell migration was promoted. IL-1 p protein levels were decreased and SHIP1 expression was downregulated. When miR155 and its target SHIP1 were both activated, the expression of both was unaltered, maintaining cell homeostasis. This points towards miR-155 overexpression can trigger inflammation, and that miR-155 and its target genes act as a molecular switch role in the development of inflammation.

Authors

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

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available