Cargando…

MircroRNA Let-7a-5p in Airway Smooth Muscle Cells is Most Responsive to High Stretch in Association With Cell Mechanics Modulation

Objective: High stretch (strain >10%) can alter the biomechanical behaviors of airway smooth muscle cells which may play important roles in diverse lung diseases such as asthma and ventilator-induced lung injury. However, the underlying modulation mechanisms for high stretch-induced mechanobiolog...

Descripción completa

Detalles Bibliográficos
Autores principales: Wen, Kang, Ni, Kai, Guo, Jia, Bu, Bing, Liu, Lei, Pan, Yan, Li, Jingjing, Luo, Mingzhi, Deng, Linhong
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8990250/
https://www.ncbi.nlm.nih.gov/pubmed/35399286
http://dx.doi.org/10.3389/fphys.2022.830406
_version_ 1784683338823368704
author Wen, Kang
Ni, Kai
Guo, Jia
Bu, Bing
Liu, Lei
Pan, Yan
Li, Jingjing
Luo, Mingzhi
Deng, Linhong
author_facet Wen, Kang
Ni, Kai
Guo, Jia
Bu, Bing
Liu, Lei
Pan, Yan
Li, Jingjing
Luo, Mingzhi
Deng, Linhong
author_sort Wen, Kang
collection PubMed
description Objective: High stretch (strain >10%) can alter the biomechanical behaviors of airway smooth muscle cells which may play important roles in diverse lung diseases such as asthma and ventilator-induced lung injury. However, the underlying modulation mechanisms for high stretch-induced mechanobiological responses in ASMCs are not fully understood. Here, we hypothesize that ASMCs respond to high stretch with increased expression of specific microRNAs (miRNAs) that may in turn modulate the biomechanical behaviors of the cells. Thus, this study aimed to identify the miRNA in cultured ASMCs that is most responsive to high stretch, and subsequently investigate in these cells whether the miRNA expression level is associated with the modulation of cell biomechanics. Methods: MiRNAs related to inflammatory airway diseases were obtained via bioinformatics data mining, and then tested with cultured ASMCs for their expression variations in response to a cyclic high stretch (13% strain) simulating in vivo ventilator-imposed strain on airways. Subsequently, we transfected cultured ASMCs with mimics and inhibitors of the miRNA that is most responsive to the high stretch, followed by evaluation of the cells in terms of morphology, stiffness, traction force, and mRNA expression of cytoskeleton/focal adhesion-related molecules. Results: 29 miRNAs were identified to be related to inflammatory airway diseases, among which let-7a-5p was the most responsive to high stretch. Transfection of cultured human ASMCs with let-7a-5p mimics or inhibitors led to an increase or decrease in aspect ratio, stiffness, traction force, migration, stress fiber distribution, mRNA expression of α-smooth muscle actin (SMA), myosin light chain kinase, some subfamily members of integrin and talin. Direct binding between let-7a-5p and ItgαV was also verified in classical model cell line by using dual-luciferase assays. Conclusion: We demonstrated that high stretch indeed enhanced the expression of let-7a-5p in ASMCs, which in turn led to changes in the cells’ morphology and biomechanical behaviors together with modulation of molecules associated with cytoskeletal structure and focal adhesion. These findings suggest that let-7a-5p regulation is an alternative mechanism for high stretch-induced effect on mechanobiology of ASMCs, which may contribute to understanding the pathogenesis of high stretch-related lung diseases.
format Online
Article
Text
id pubmed-8990250
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-89902502022-04-09 MircroRNA Let-7a-5p in Airway Smooth Muscle Cells is Most Responsive to High Stretch in Association With Cell Mechanics Modulation Wen, Kang Ni, Kai Guo, Jia Bu, Bing Liu, Lei Pan, Yan Li, Jingjing Luo, Mingzhi Deng, Linhong Front Physiol Physiology Objective: High stretch (strain >10%) can alter the biomechanical behaviors of airway smooth muscle cells which may play important roles in diverse lung diseases such as asthma and ventilator-induced lung injury. However, the underlying modulation mechanisms for high stretch-induced mechanobiological responses in ASMCs are not fully understood. Here, we hypothesize that ASMCs respond to high stretch with increased expression of specific microRNAs (miRNAs) that may in turn modulate the biomechanical behaviors of the cells. Thus, this study aimed to identify the miRNA in cultured ASMCs that is most responsive to high stretch, and subsequently investigate in these cells whether the miRNA expression level is associated with the modulation of cell biomechanics. Methods: MiRNAs related to inflammatory airway diseases were obtained via bioinformatics data mining, and then tested with cultured ASMCs for their expression variations in response to a cyclic high stretch (13% strain) simulating in vivo ventilator-imposed strain on airways. Subsequently, we transfected cultured ASMCs with mimics and inhibitors of the miRNA that is most responsive to the high stretch, followed by evaluation of the cells in terms of morphology, stiffness, traction force, and mRNA expression of cytoskeleton/focal adhesion-related molecules. Results: 29 miRNAs were identified to be related to inflammatory airway diseases, among which let-7a-5p was the most responsive to high stretch. Transfection of cultured human ASMCs with let-7a-5p mimics or inhibitors led to an increase or decrease in aspect ratio, stiffness, traction force, migration, stress fiber distribution, mRNA expression of α-smooth muscle actin (SMA), myosin light chain kinase, some subfamily members of integrin and talin. Direct binding between let-7a-5p and ItgαV was also verified in classical model cell line by using dual-luciferase assays. Conclusion: We demonstrated that high stretch indeed enhanced the expression of let-7a-5p in ASMCs, which in turn led to changes in the cells’ morphology and biomechanical behaviors together with modulation of molecules associated with cytoskeletal structure and focal adhesion. These findings suggest that let-7a-5p regulation is an alternative mechanism for high stretch-induced effect on mechanobiology of ASMCs, which may contribute to understanding the pathogenesis of high stretch-related lung diseases. Frontiers Media S.A. 2022-03-25 /pmc/articles/PMC8990250/ /pubmed/35399286 http://dx.doi.org/10.3389/fphys.2022.830406 Text en Copyright © 2022 Wen, Ni, Guo, Bu, Liu, Pan, Li, Luo and Deng. https://creativecommons.org/licenses/by/4.0/This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Physiology
Wen, Kang
Ni, Kai
Guo, Jia
Bu, Bing
Liu, Lei
Pan, Yan
Li, Jingjing
Luo, Mingzhi
Deng, Linhong
MircroRNA Let-7a-5p in Airway Smooth Muscle Cells is Most Responsive to High Stretch in Association With Cell Mechanics Modulation
title MircroRNA Let-7a-5p in Airway Smooth Muscle Cells is Most Responsive to High Stretch in Association With Cell Mechanics Modulation
title_full MircroRNA Let-7a-5p in Airway Smooth Muscle Cells is Most Responsive to High Stretch in Association With Cell Mechanics Modulation
title_fullStr MircroRNA Let-7a-5p in Airway Smooth Muscle Cells is Most Responsive to High Stretch in Association With Cell Mechanics Modulation
title_full_unstemmed MircroRNA Let-7a-5p in Airway Smooth Muscle Cells is Most Responsive to High Stretch in Association With Cell Mechanics Modulation
title_short MircroRNA Let-7a-5p in Airway Smooth Muscle Cells is Most Responsive to High Stretch in Association With Cell Mechanics Modulation
title_sort mircrorna let-7a-5p in airway smooth muscle cells is most responsive to high stretch in association with cell mechanics modulation
topic Physiology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8990250/
https://www.ncbi.nlm.nih.gov/pubmed/35399286
http://dx.doi.org/10.3389/fphys.2022.830406
work_keys_str_mv AT wenkang mircrornalet7a5pinairwaysmoothmusclecellsismostresponsivetohighstretchinassociationwithcellmechanicsmodulation
AT nikai mircrornalet7a5pinairwaysmoothmusclecellsismostresponsivetohighstretchinassociationwithcellmechanicsmodulation
AT guojia mircrornalet7a5pinairwaysmoothmusclecellsismostresponsivetohighstretchinassociationwithcellmechanicsmodulation
AT bubing mircrornalet7a5pinairwaysmoothmusclecellsismostresponsivetohighstretchinassociationwithcellmechanicsmodulation
AT liulei mircrornalet7a5pinairwaysmoothmusclecellsismostresponsivetohighstretchinassociationwithcellmechanicsmodulation
AT panyan mircrornalet7a5pinairwaysmoothmusclecellsismostresponsivetohighstretchinassociationwithcellmechanicsmodulation
AT lijingjing mircrornalet7a5pinairwaysmoothmusclecellsismostresponsivetohighstretchinassociationwithcellmechanicsmodulation
AT luomingzhi mircrornalet7a5pinairwaysmoothmusclecellsismostresponsivetohighstretchinassociationwithcellmechanicsmodulation
AT denglinhong mircrornalet7a5pinairwaysmoothmusclecellsismostresponsivetohighstretchinassociationwithcellmechanicsmodulation