Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters











Database
Language
Publication year range
1.
Int J Mol Sci ; 23(14)2022 Jul 13.
Article in English | MEDLINE | ID: mdl-35887096

ABSTRACT

Airway mucociliary regeneration and function are key players for airway defense and are impaired in chronic obstructive pulmonary disease (COPD). Using transcriptome analysis in COPD-derived bronchial biopsies, we observed a positive correlation between cilia-related genes and microRNA-449 (miR449). In vitro, miR449 was strongly increased during airway epithelial mucociliary differentiation. In vivo, miR449 was upregulated during recovery from chemical or infective insults. miR0449-/- mice (both alleles are deleted) showed impaired ciliated epithelial regeneration after naphthalene and Haemophilus influenzae exposure, accompanied by more intense inflammation and emphysematous manifestations of COPD. The latter occurred spontaneously in aged miR449-/- mice. We identified Aurora kinase A and its effector target HDAC6 as key mediators in miR449-regulated ciliary homeostasis and epithelial regeneration. Aurora kinase A is downregulated upon miR449 overexpression in vitro and upregulated in miR449-/- mouse lungs. Accordingly, imaging studies showed profoundly altered cilia length and morphology accompanied by reduced mucociliary clearance. Pharmacological inhibition of HDAC6 rescued cilia length and coverage in miR449-/- cells, consistent with its tubulin-deacetylating function. Altogether, our study establishes a link between miR449, ciliary dysfunction, and COPD pathogenesis.


Subject(s)
Aurora Kinase A/metabolism , Histone Deacetylase 6/metabolism , MicroRNAs , Pulmonary Disease, Chronic Obstructive , Animals , Aurora Kinase A/genetics , Cilia/genetics , Epithelial Cells , Mice , MicroRNAs/genetics , Pulmonary Disease, Chronic Obstructive/genetics , Tubulin/genetics
2.
Cells ; 11(3)2022 02 02.
Article in English | MEDLINE | ID: mdl-35159335

ABSTRACT

We recently identified microRNAs (miRNAs) associated with chronic mucus hypersecretion (CMH) in chronic obstructive pulmonary disease (COPD), which were expressed in both airway epithelial cells and fibroblasts. We hypothesized that these miRNAs are involved in communication between fibroblasts and epithelium, contributing to airway remodeling and CMH in COPD. Primary bronchial epithelial cells (PBECs) differentiated at the air-liquid interface, and airway fibroblasts (PAFs) from severe COPD patients with CMH were cultured alone or together. RNA was isolated and miRNA expression assessed. miRNAs differentially expressed after co-culturing were studied functionally using overexpression with mimics in mucus-expressing human lung A549 epithelial cells or normal human lung fibroblasts. In PBECs, we observed higher miR-708-5pexpression upon co-culture with fibroblasts, and miR-708-5p expression decreased upon mucociliary differentiation. In PAFs, let-7a-5p, miR-31-5p and miR-146a-5p expression was significantly increased upon co-culture. miR-708-5p overexpression suppressed mucin 5AC (MUC5AC) secretion in A549, while let-7a-5poverexpression suppressed its target gene COL4A1 in lung fibroblasts. Our findings suggest that let-7a-5p, miR-31-5p and miR-146a-5p may be involved in CMH via fibroblasts-epithelium crosstalk, including extracellular matrix gene regulation, while airway epithelial expression of miR-708-5p may be involved directly, regulating mucin production. These findings shed light on miRNA-mediated mechanisms underlying CMH, an important symptom in COPD.


Subject(s)
MicroRNAs , Pulmonary Disease, Chronic Obstructive , Epithelium/metabolism , Fibroblasts/metabolism , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Mucus/metabolism , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/metabolism
4.
Curr Opin Pulm Med ; 25(1): 87-93, 2019 01.
Article in English | MEDLINE | ID: mdl-30394902

ABSTRACT

PURPOSE OF REVIEW: Numerous signaling pathways and inflammatory responses in cells and tissues are under microRNA (miRNA) control. In the present review, the role of miRNAs and exosomes in the pathogenesis of asthma will be discussed. RECENT FINDINGS: MiRNAs differentially expressed with asthma, for example, miRNA-34/449, let-7, miRNA-19, miRNA-21, and miRNA-455, were identified in various cell types and tissues including epithelial cells, T cells, type 2 innate lymphoid cells, lung tissues, and smooth muscles. Current data suggest the involvement of these miRNAs in epithelial differentiation, mucus production, airway remodeling, inflammation, etc. However, it is often difficult to predict which genes are targeted by a specific miRNA. We recently combined genome-wide miRNA analyses together with transcriptome in bronchial biopsies, in relation to chronic mucus hypersecretion, then performed a genome-wide miRNA-mRNA network analysis and identified the key miRNA regulators for chronic mucus hypersecretion. SUMMARY: There is now growing evidence suggesting that miRNAs play critically important roles in asthma. Several asthma-associated miRNAs have already been identified. Although miRNAs are attractive targets for therapeutic intervention, a safe and effective delivery to target tissues and cells in humans remains a challenge.


Subject(s)
Asthma/genetics , Asthma/metabolism , Exosomes/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Mucus/metabolism , Asthma/drug therapy , Gene Expression Profiling , Genome-Wide Association Study , Humans , RNA, Messenger/analysis , Transcriptome
5.
Eur Respir J ; 52(3)2018 09.
Article in English | MEDLINE | ID: mdl-30072506

ABSTRACT

Chronic mucus hypersecretion (CMH) is a common feature in chronic obstructive pulmonary disease (COPD) and is associated with worse prognosis and quality of life. This study aimed to identify microRNA (miRNA)-mRNA regulatory networks underlying CMH.The expression profiles of miRNA and mRNA in bronchial biopsies from 63 COPD patients were associated with CMH using linear regression. Potential mRNA targets of each CMH-associated miRNA were identified using Pearson correlations. Gene set enrichment analysis (GSEA) and STRING (search tool for the retrieval of interacting genes/proteins) analysis were used to identify key genes and pathways.20 miRNAs and 539 mRNAs were differentially expressed with CMH in COPD. The expression of 10 miRNAs was significantly correlated with the expression of one or more mRNAs. Of these, miR-134-5p, miR-146a-5p and the let-7 family had the highest representation of CMH-associated mRNAs among their negatively correlated predicted targets. KRAS and EDN1 were identified as key regulators of CMH and were negatively correlated predicted targets of miR-134-5p and let-7a-5p, let-7d-5p, and let-7f-5p, respectively. GSEA suggested involvement of MUC5AC-related genes and several other relevant gene sets in CMH. The lower expression of miR-134-5p was confirmed in primary airway fibroblasts from COPD patients with CMH.We identified miR-134-5p, miR-146a-5p and let-7 family, along with their potential target genes including KRAS and EDN1, as potential key miRNA-mRNA networks regulating CMH in COPD.


Subject(s)
MicroRNAs/genetics , Pulmonary Disease, Chronic Obstructive/genetics , Respiratory Mucosa/metabolism , Aged , Bronchi/pathology , Female , Gene Expression Profiling , Gene Regulatory Networks , Humans , Linear Models , Male , Middle Aged , Proto-Oncogene Proteins p21(ras)/genetics , Pulmonary Disease, Chronic Obstructive/physiopathology , Quality of Life , RNA, Messenger/genetics
6.
Eur Respir J ; 49(5)2017 05.
Article in English | MEDLINE | ID: mdl-28546273

ABSTRACT

We previously reported that epithelial-derived interleukin (IL)-1α drives fibroblast-derived inflammation in the lung epithelial-mesenchymal trophic unit. Since miR-146a-5p has been shown to negatively regulate IL-1 signalling, we investigated the role of miR-146a-5p in the regulation of IL-1α-driven inflammation in chronic obstructive pulmonary disease (COPD).Human bronchial epithelial (16HBE14o-) cells were co-cultured with control and COPD-derived primary human lung fibroblasts (PHLFs), and miR-146a-5p expression was assessed with and without IL-1α neutralising antibody. Genomic DNA was assessed for the presence of the single nucleotide polymorphism (SNP) rs2910164. miR-146a-5p mimics were used for overexpression studies to assess IL-1α-induced signalling and IL-8 production by PHLFs.Co-culture of PHLFs with airway epithelial cells significantly increased the expression of miR-146a-5p and this induction was dependent on epithelial-derived IL-1α. miR-146a-5p overexpression decreased IL-1α-induced IL-8 secretion in PHLFs via downregulation of IL-1 receptor-associated kinase-1. In COPD PHLFs, the induction of miR-146a-5p was significantly less compared with controls and was associated with the SNP rs2910164 (GG allele) in the miR-146a-5p gene.Our results suggest that induction of miR-146a-5p is involved in epithelial-fibroblast communication in the lungs and negatively regulates epithelial-derived IL-1α induction of IL-8 by fibroblasts. The decreased levels of miR-146a-5p in COPD fibroblasts may induce a more pro-inflammatory phenotype, contributing to chronic inflammation in COPD.


Subject(s)
Epithelium/metabolism , Fibroblasts/metabolism , MicroRNAs/metabolism , Pulmonary Disease, Chronic Obstructive/metabolism , Alleles , Antibodies, Neutralizing/chemistry , Bronchi/metabolism , Cell Line, Tumor , Cigarette Smoking , Coculture Techniques , Culture Media, Conditioned , Epithelial Cells/metabolism , Humans , Inflammation , Interleukin-1alpha/metabolism , Interleukin-8/metabolism , Polymorphism, Single Nucleotide , Signal Transduction , Tobacco Products
7.
Aging (Albany NY) ; 8(8): 1608-35, 2016 08.
Article in English | MEDLINE | ID: mdl-27385366

ABSTRACT

Senescent cancer-associated fibroblasts (CAF) develop a senescence-associated secretory phenotype (SASP) that is believed to contribute to cancer progression. The mechanisms underlying SASP development are, however, poorly understood. Here we examined the functional role of microRNA in the development of the SASP in normal fibroblasts and CAF. We identified a microRNA, miR-335, up-regulated in the senescent normal fibroblasts and CAF and able to modulate the secretion of SASP factors and induce cancer cell motility in co-cultures, at least in part by suppressing the expression of phosphatase and tensin homologue (PTEN). Additionally, elevated levels of cyclo-oxygenase 2 (PTGS2; COX-2) and prostaglandin E2 (PGE2) secretion were observed in senescent fibroblasts, and inhibition of COX-2 by celecoxib reduced the expression of miR-335, restored PTEN expression and decreased the pro-tumourigenic effects of the SASP. Collectively these data demonstrate the existence of a novel miRNA/PTEN-regulated pathway modulating the inflammasome in senescent fibroblasts.


Subject(s)
Cancer-Associated Fibroblasts/metabolism , Cellular Senescence/physiology , Cyclooxygenase 2/metabolism , MicroRNAs/metabolism , PTEN Phosphohydrolase/metabolism , Signal Transduction/physiology , Cancer-Associated Fibroblasts/drug effects , Celecoxib/pharmacology , Cell Movement/drug effects , Cell Movement/physiology , Coculture Techniques , Dinoprostone/metabolism , Humans , Phenotype , Signal Transduction/drug effects , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL