Citation

  • Authors: Naidu, S., Shi, L., Magee, P., Middleton, J. D., Lagana, A., Sahoo, S., Leong, H. S., Galvin, M., Frese, K., Dive, C., Guzzardo, V., Fassan, M., Garofalo, M.
  • Year: 2017
  • Journal: Sci Rep 7 15441
  • Applications: in vivo / miRNA / in vivo-jetPEI

Method

When the tumours reached 200 mm^3, mice (n = 4) were randomized by sequential assignment to miR-Ctr, miR-27b/24-1 and miR-125a-5p treatment groups and injected with in vivo-jetPEI complexed with 100 μg of each microRNA four times in three weeks.

Abstract

In NSCLC alterations in PDGF receptors are markers of worst prognosis and efficient targeting of these receptors is yet to be achieved. In this study, we explored PDGFR-regulated microRNAs demonstrating that miR-23b cluster and miR-125a-5p are downregulated by increased expression of PDGFR-alpha or PDGFR-beta in NSCLC cells. Mechanistically, the expression of these microRNAs is positively regulated by p53 and negatively modulated by NF-kB p65. Forced expression of miR-23b cluster or miR-125a-5p enhanced drug sensitivity and suppressed invasiveness of NSCLC cells by silencing several genes involved in oncogenic KRAS and NF-kB pathways, including SOS1, GRB2, IQGAP1, RALA, RAF-1, IKKbeta, AKT2, ERK2 and KRAS itself. Of note, an inverse correlation between miR-23b cluster, miR-125a-5p and respective target genes was also found in vivo in a large dataset of lung adenocarcinoma samples. Furthermore, in vivo delivery of miR-23b cluster or miR-125a-5p significantly repressed tumour growth in a highly aggressive NSCLC circulating tumour cell (CTC) patient derived explant (CDX) mouse model. In conclusion, our finding sheds light on the PDGFR signaling and endorses the possibility to employ miR-23b cluster and miR-125a-5p as therapeutic tools to silence simultaneously a range of redundant pathways and main effectors of tumorigenesis in NSCLC.

Go to