Cargando…

MicroRNA-133 Targets Phosphodiesterase 1C in Drosophila and Human Oral Cancer Cells to Regulate Epithelial-Mesenchymal Transition

Non-coding microRNAs (miRNAs) have been proposed to play diverse roles in cancer biology, including epithelial-mesenchymal transition (EMT) crucial for cancer progression. Previous comparative studies revealed distinct expression profiles of miRNAs relevant to tumorigenesis and progression of oral c...

Descripción completa

Detalles Bibliográficos
Autores principales: Jung, Ji Eun, Lee, Joo Young, Park, Hae Ryoun, Kang, Ji Wan, Kim, Yun Hak, Lee, Ji Hye
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Ivyspring International Publisher 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8317528/
https://www.ncbi.nlm.nih.gov/pubmed/34335946
http://dx.doi.org/10.7150/jca.56138
_version_ 1783730089035300864
author Jung, Ji Eun
Lee, Joo Young
Park, Hae Ryoun
Kang, Ji Wan
Kim, Yun Hak
Lee, Ji Hye
author_facet Jung, Ji Eun
Lee, Joo Young
Park, Hae Ryoun
Kang, Ji Wan
Kim, Yun Hak
Lee, Ji Hye
author_sort Jung, Ji Eun
collection PubMed
description Non-coding microRNAs (miRNAs) have been proposed to play diverse roles in cancer biology, including epithelial-mesenchymal transition (EMT) crucial for cancer progression. Previous comparative studies revealed distinct expression profiles of miRNAs relevant to tumorigenesis and progression of oral cancer. With putative targets of these miRNAs mostly validated in vitro, it remains unclear whether similar miRNA-target relationships exist in vivo. In this study, we employed a hybrid approach, utilizing both Drosophila melanogaster and human oral cancer cells, to validate projected miRNA-target relationships relevant to EMT. Notably, overexpression of dme-miR-133 resulted in significant tissue growth in Drosophila larval wing discs. The RT-PCR analysis successfully validated a subset of its putative targets, including Pde1c. Subsequent experiments performed in oral cancer cells confirmed conserved targeting of human PDE1C by hsa-miR-133. Furthermore, the elevated level of miR-133 and its targeting of PDE1C was positively correlated with enhanced migrative ability of oral cancer cells treated with LPS, along with the molecular signature of a facilitated EMT process induced by LPS and TGF-β. The analysis on the RNAseq data also revealed a negative correlation between the expression level of hsa-miR-133 and the survival of oral cancer patients. Taken together, our mammal-to-Drosophila-to-mammal approach successfully validates targeting of PDE1C by miR-133 both in vivo and in vitro, underlying the promoted EMT phenotypes and potentially influencing the prognosis of oral cancer patients. This hybrid approach will further aid to widen our scope in investigation of intractable human malignancies, including oral cancer.
format Online
Article
Text
id pubmed-8317528
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher Ivyspring International Publisher
record_format MEDLINE/PubMed
spelling pubmed-83175282021-07-29 MicroRNA-133 Targets Phosphodiesterase 1C in Drosophila and Human Oral Cancer Cells to Regulate Epithelial-Mesenchymal Transition Jung, Ji Eun Lee, Joo Young Park, Hae Ryoun Kang, Ji Wan Kim, Yun Hak Lee, Ji Hye J Cancer Research Paper Non-coding microRNAs (miRNAs) have been proposed to play diverse roles in cancer biology, including epithelial-mesenchymal transition (EMT) crucial for cancer progression. Previous comparative studies revealed distinct expression profiles of miRNAs relevant to tumorigenesis and progression of oral cancer. With putative targets of these miRNAs mostly validated in vitro, it remains unclear whether similar miRNA-target relationships exist in vivo. In this study, we employed a hybrid approach, utilizing both Drosophila melanogaster and human oral cancer cells, to validate projected miRNA-target relationships relevant to EMT. Notably, overexpression of dme-miR-133 resulted in significant tissue growth in Drosophila larval wing discs. The RT-PCR analysis successfully validated a subset of its putative targets, including Pde1c. Subsequent experiments performed in oral cancer cells confirmed conserved targeting of human PDE1C by hsa-miR-133. Furthermore, the elevated level of miR-133 and its targeting of PDE1C was positively correlated with enhanced migrative ability of oral cancer cells treated with LPS, along with the molecular signature of a facilitated EMT process induced by LPS and TGF-β. The analysis on the RNAseq data also revealed a negative correlation between the expression level of hsa-miR-133 and the survival of oral cancer patients. Taken together, our mammal-to-Drosophila-to-mammal approach successfully validates targeting of PDE1C by miR-133 both in vivo and in vitro, underlying the promoted EMT phenotypes and potentially influencing the prognosis of oral cancer patients. This hybrid approach will further aid to widen our scope in investigation of intractable human malignancies, including oral cancer. Ivyspring International Publisher 2021-07-03 /pmc/articles/PMC8317528/ /pubmed/34335946 http://dx.doi.org/10.7150/jca.56138 Text en © The author(s) https://creativecommons.org/licenses/by/4.0/This is an open access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/). See http://ivyspring.com/terms for full terms and conditions.
spellingShingle Research Paper
Jung, Ji Eun
Lee, Joo Young
Park, Hae Ryoun
Kang, Ji Wan
Kim, Yun Hak
Lee, Ji Hye
MicroRNA-133 Targets Phosphodiesterase 1C in Drosophila and Human Oral Cancer Cells to Regulate Epithelial-Mesenchymal Transition
title MicroRNA-133 Targets Phosphodiesterase 1C in Drosophila and Human Oral Cancer Cells to Regulate Epithelial-Mesenchymal Transition
title_full MicroRNA-133 Targets Phosphodiesterase 1C in Drosophila and Human Oral Cancer Cells to Regulate Epithelial-Mesenchymal Transition
title_fullStr MicroRNA-133 Targets Phosphodiesterase 1C in Drosophila and Human Oral Cancer Cells to Regulate Epithelial-Mesenchymal Transition
title_full_unstemmed MicroRNA-133 Targets Phosphodiesterase 1C in Drosophila and Human Oral Cancer Cells to Regulate Epithelial-Mesenchymal Transition
title_short MicroRNA-133 Targets Phosphodiesterase 1C in Drosophila and Human Oral Cancer Cells to Regulate Epithelial-Mesenchymal Transition
title_sort microrna-133 targets phosphodiesterase 1c in drosophila and human oral cancer cells to regulate epithelial-mesenchymal transition
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8317528/
https://www.ncbi.nlm.nih.gov/pubmed/34335946
http://dx.doi.org/10.7150/jca.56138
work_keys_str_mv AT jungjieun microrna133targetsphosphodiesterase1cindrosophilaandhumanoralcancercellstoregulateepithelialmesenchymaltransition
AT leejooyoung microrna133targetsphosphodiesterase1cindrosophilaandhumanoralcancercellstoregulateepithelialmesenchymaltransition
AT parkhaeryoun microrna133targetsphosphodiesterase1cindrosophilaandhumanoralcancercellstoregulateepithelialmesenchymaltransition
AT kangjiwan microrna133targetsphosphodiesterase1cindrosophilaandhumanoralcancercellstoregulateepithelialmesenchymaltransition
AT kimyunhak microrna133targetsphosphodiesterase1cindrosophilaandhumanoralcancercellstoregulateepithelialmesenchymaltransition
AT leejihye microrna133targetsphosphodiesterase1cindrosophilaandhumanoralcancercellstoregulateepithelialmesenchymaltransition