SciELO - Scientific Electronic Library Online

 
vol.85 número1Uses of magnesium sulfate in anesthesiologyDysphagia rehabilitation in post-COVID patients: Review of the literature índice de autoresíndice de assuntospesquisa de artigos
Home Pagelista alfabética de periódicos  

Serviços Personalizados

Journal

Artigo

Indicadores

Links relacionados

  • Não possue artigos similaresSimilares em SciELO

Compartilhar


Revista médica del Hospital General de México

versão On-line ISSN 2524-177Xversão impressa ISSN 0185-1063

Rev. med. Hosp. Gen. Méx. vol.85 no.1 Ciudad de México Jan./Mar. 2022  Epub 02-Maio-2022

https://doi.org/10.24875/hgmx.21000049 

Review articles

MiRNAs involved in the signaling pathways associated to the pathogenesis of idiopathic pulmonary fibrosis

Ana R. Cadena-Suárez1  * 

1Molecular Biology Laboratory “Ismael Cosío Villegas” National Institute of Respiratory Diseases, Mexico City, Mexico


Abstract

Idiopathic Pulmonary Fibrosis (IPF) is a progressive and multifactorial interstitial lung disease whose pathophysiology remains unclear. The process initiates by repeated epithelial lung injuries followed by basal membrane destruction occasioning the activation of the epithelial mesenchymal transition (EMT) and myofibroblasts which carry out an excessive synthesis of extracellular matrix (ECM) proteins. Several studies have associated microRNAs (miRNAs) to the biogenesis and development of IPF because miRNAs participate in the processes of apoptosis, proliferation, differentiation and interaction between cells thanks to their role as activators or inhibitors of different receptors in the signaling pathways of TGF-β, Wnt/β-catenin and PI3K-Akt-FOXO3a-mTOR, which are the main and most studied pathways involved in lung fibrosis. In this context, the knowledge on the altered miRNAs expression and even more their role in the different signaling pathways in IPF will shed light on new therapeutic targets for this lung disease.

Keywords miRNAs; Idiopathic pulmonary fibrosis; Epithelial mesenchymal transition; Myofibroblasts

Introduction

Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive disease with poor prognosis, and f ew therapeutic options. The incidence and prevalence increase with age, and it is most common seen in male patients older than 65 years. At first, IPF was considered as a chronic inflammatory disorder characterized by progressive fibrosis, however, evidence indicate that this pathology is a product of an epithelial-driven disorder associated with environmental, genetic risk factors, aging-associated processes and a profibrotic epigenetic reprogramming1. During the development of IPF, lung parenchyma shows repeated epithelial cell injuries with an aberrant reparation mechanism that cause several lung scars, mainly caused by an excessive fibroblasts activity and their trans differentiation to myofibroblasts. Moreover, the epithelial-mesenchymal transition (EMT) promotes myofibroblasts accumulation, and synthesis of an excessive amount of collagen and extracellular matrix (ECM) proteins by the alveolar epithelial cells (AECs)2.

MiRNAs are short, non-coding RNAs with an extension of approximately 21-23 nucleotides which play a post-transcriptional regulatory role by targeting specific messenger RNAs for degradation and/or translational repression. To date, there are already some reports about profibrotic and anti-fibrotic miRNAs dysregulation in the pathogenesis of IPF. These mechanisms are carried out since miRNAs have overlapping functions because they can either regulate a single gene or several genes can be affected by a single miRNA. For this reason, is relevant to update the information about altered miRNAs involved in the crucial signaling pathways implicated in IPF to have a better understanding of their pathogenesis mechanisms and to propose possible potential therapeutic targets3.

Materials and Methods

This review is focused on a PubMed searching using the following key words: “miRNAs involved in IPF”, “miRNAs involved in cellular signaling pathways in IPF” and “miRNAs involved in the TGF-β pathway”. The selection was reduced to articles written in the years 2016 to 2020 in English language. The articles found were around fifty; however, the selected papers were the ones that related miRNAs with the TGF-β, Wnt/β-catenin and PI3K-Akt-FOXO3a-mTOR signaling pathways since these signaling pathways are the most studied ones in pulmonary fibrosis process.

For better understanding, the manuscript is organized according to miRNAs participation in each signaling pathway involved in the pathogenesis in IPF.

First, a general summary about the main signaling pathways involved in the pathogenesis of IPF is explained and then there is a description of the function of each miRNA within them.

EMT process in IPF

EMT is a process in which epithelial cells gradually lose their cell-to-cell junctions, adherens junctions and apical-basal polarity to acquire a mesenchymal cell phenotype. This phenotype gains new invasive properties, and re-synthesizing new extracellular matrix proteins. The EMT process, can be activated by transcription factors and by signaling pathways, being the transforming growth factor beta 1 (TGF-β) its best inductor4. This process is controlled by three families of transcription factors: the zinc finger Snail (Snail/Slug) and zinc finger E-Box binding homeobox 1 and 2 (ZEB1/ZEB2) which have, as main function, the repression of E-cadherin expression5. Nowadays, it is well stablished that EMT process in IPF is regulated also, by epigenetic modulators like miRNAs which can act by activating or reverting lung fibrosis through the regulation of targets expression involved in EMT signaling pathways such as TGF-β, Wnt/β-catenin and PI3K-Akt-FOXO3a-mTOR, which induce the activation of resident fibroblasts and their trans differentiation into myofibroblasts, for example, miRNA-326 reduces the expression of profibrotic genes by targetting TGF-β and miRNA-26 directly downregulates CTGF which is the responsable for collagen formation6.

Signaling pathways involved in EMT process

EMT is regulated by extracellular ligands such as TGF-β, interleukin-1 (IL-1), connective tissue growth factor (CTGF) and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kb), which is activated by tumor necrosis factor alpha (TNF-a), insulin-like growth factor-1 (IGF-1), secreted glycoproteins of the wingless/integrate-1 (Wnt)5 and ADAM metallopeptidase with thrombospondin type motif 1 (ADAMTS1)7.

All of them, have the property of initiating the intracellular signaling cascades through the activation of one or more EMT-driving transcription factors such as SNAIL1/2 and ZEB1/2 and downregulating the expression of adhesion molecules like E-cadherin, being the high-mobility group AT-hook2 (HMGA2) the one that facilitates the transcription of SNAIL15.

It has also been proved that MiRNAs have a stimulant role in extracellular matrix protein synthesis through collagen genes such as COL1A1, COL1A2, COL3A1, COL5A2, COL4A2, LOXL and matrix metallopeptidase (MMP) like MMP2, MMP3 and MMP10, among others8.

TGF-ß signaling pathway

In lung diseases like IPF, there is an increasing evidence that proves the activation of TGF-β pathway by lung cells to maintain homeostasis in response to tissue injury9. This cytokine is secreted, in a latent form, bound to latency-associated peptide (LAP), but when it is released from this peptide, TFG-b dimers conform a complex with the type I TGF-β receptor (TGF-βRI) and type II receptor (TGF-βRII). This complex initiates a signal transduction by a Smad-dependent or non-Smad-dependent pathway8. In the Smad-dependent pathway, activated TGF-βRI phosphorylates cytoplasmic Smad2/3 transcription factors and translocate them into the nucleus. Smad4 facilitate this process and together with ZEB1/2 complex modulate the pro-fibrotic genes expression associated with EMT process, fibroblasts differentiation, proliferation migration and deposition of collagen10. Moreover, the Smad 2/3 complex represses the expression of E-cadherin through SNAIL1 and SNAIL2 which induce the expression of mesenchymal proteins such as N-cadherin, fibronectin, and metalloproteinases11. On the other hand, Smad6 and Smad7 inhibits the phosphorylation of Smad 2/3 complex by competing with Smad4 or TGF-βRI which is also regulated by Smurf2 E3 ubiquitin ligase. For this way, TGF-β signaling pathway is downregulated by Smad6, Smad7, and Smurf210.

In the non-Smad mediated pathway TGF-β can activate other signaling cascades, like mitogen-activated protein kinase (MAPK) pathway which can mediate Smad phosphorylation and activation through the activation of extracellular signal-regulated kinase (Erk) and c-Jun N-terminal kinase (JNK) pathways. Also, TGF-β can activate other signaling mediators such as p38 MAPK kinase kinases family (MAPKKKS) and TGF-β activated kinase 1 (TAK1) which has the function of activating NF-kB signaling pathway. This dual ability of TGF-β to activate Smad and non Smad signaling pathways has an important role in the EMT process12.

There are many others signaling pathways involved in the pathogenesis of IPF, however, the Wnt/β-catenin and phosphoinositide 3 kinase (PI3K)/protein kinase B (Akt) (PI3K/Akt) signaling pathways are the most relevant because they are related to cell proliferation, differentiation, and extracellular matrix production.

Wnt/ß catenin signaling pathway

In presence of injury Wnt/β-catenin signaling pathway promotes alveolar repair and remodeling, fibroblast migration and proliferation and extracellular matrix production through the stimulation of β-catenin by Wnt13.

The activation of this pathway begins with the stimulation of TGF-β which generates the release of Wnt from the axis inhibition protein 1 (AXIN1)- glycogen synthase kinase 3 beta (GSK3b) - adenomatous polyposis coli (APC) (AXIN1/GSK3b/APC) complex, which, in a steady state, leads to ubiquitin-mediated proteolysis of β-catenin. When Wnt is released from this complex, there is a nuclear translocation of β-catenin via the phosphorylation of extracellular signal-regulated kinases 1 and 2 (ERK1/2) and suppression of GSK-3b and AXIN1 activity, this causes the accumulation of β-catenin and then suffers a nuclear translocation, by this way it can regulate the fibronectin gene 1 (FN1), the metalloproteinase 7 (Mmp 7) and cyclin D1 with the consequent production of extracellular matrix14.

PI3K-Akt signaling pathway

This pathway has the function of regulating fibroblasts growth, proliferation, apoptosis, and collagen production; however, this pathway is down regulated in IPF which leads to Akt up regulation under mechanisms that modulate the activity of the phosphatidylinositol 3-kinase (PI3K)-protein kinase B (Akt)- forkhead box O3 (FoxO3a) and mammalian target of rapamycin (mTOR) (PI3K-Akt-FOXO3a-mTOR) axis. PI3K stimulates the synthesis of phosphatidylinositol-3,4,5-triphosphate (PIP3) which causes the activation of Akt. One of Akt targets is FoxO3a which is a powerful inhibitor of cell cycle due to the activation of the cyclin-dependent kinases (CDK) inhibitor protein p27 which promotes G1 arrest, however, FoxO3a activity is abnormally low in IPF fibroblasts because phosphatase and tensin homolog (PTEN), the main regulator of this pathway, is poorly expressed. This leads to fibroblasts autophagy inhibition thus promoting fibroblasts proliferation, increases the expression of alpha-smooth muscle actin (a-SMA) and fibronectin and reduces collagen degradation creating a collagen-rich environment15.

MTOR has a role in cell growth, proliferation and apoptosis, it forms two distinct complexes which are mammalian target of rapamycin complex 1 (mTORC1) and mammalian target of rapamycin complex 2 (mTORC2), the first one controls cell growth and the second one regulates cell proliferation and survival: being the tuberous Sclerosis complex (TSC) the negative regulator of mTORC1 and PI3K the negative regulator of mTORC216.

MiRNAs have an important role in these signaling pathways because they can act as pro fibrotic or antifibrotic mediators through the stimulation or inactivation of different receptors in the signaling cascade, further examples of this will be seen below.

Figure 1 summarizes the different signaling pathways mentioned above, such as TGF-β, Wnt/β-catenin and PI3K-Akt-FOXO3a-mTOR as well as their cross-linking. Also, several receptors involved in these pathways are illustrated, which can be stimulated or inhibited by the action of different miRNAs which can either stimulate or inhibit the trans differentiation of fibroblasts into myofibroblasts, pro fibrotic genes activation and extracellular matrix proteins synthesis.

Figure 1 Proposal crosstalk between TGF-β with Smad, non-Smad dependent pathway, Wnt/β-catenin and PI3K-Akt-FOXO3a-mTOR signaling pathways involved in the EMT process and some examples of miRNAs mentioned in this review involved in the fibrotic process. (Image inspired in the work of Derynck, R., & Zhang, Y.E. [2003], Rajasekaran, S., Rajaguru, P., & Sudhakar Gandhi, P. S. [2015] and Razali, R. A., Lokanathan, Y., Yazid, M. D., Ansari, A. S., Saim, A. B., & Hj Idrus, R. B. [2019]). 

MiRNAs involved in the pathogenesis of IPF

The compilation of miRNAs implicated in IPF was grouped depending on the signaling route and the process they regulate. They are better illustrated in Table 1.

Table 1 MiRNAs involved in the crucial signaling pathways of Idiopathic Pulmonary Fibrosis 

Sample miRNA Function in health Target of action Effect Status in IPF Reference
MiRNAs involved in the EMT in a Smad-dependent pathway
I Bleomycin murine model miRNA-877-3p Anti-fibrotic Smad7 Regulation of fibroblasts differentiation into myofibroblasts Up regulated Wang et al., 2016
II Human A549 and bronchial epithelial cells miRNA-1343 Anti-fibrotic TGFBRI and TGFBRII Inhibits Smad2/3 phosphorylation, nuclear translocation, and reduces expression of markers of fibrosis such as α-SMA and COL1A1 Up regulated Stolzenburg et al., 2016
III Mice fibroblasts NIH3T3 miRNA-133a Anti- fibrotic COL1A1 gene and CTGF Regulation of TGF-β1induced myofibroblast differentiation Up regulated Wei et al., 2019
IV A549 cells miRNA-184 Anti-fibrotic AKT and Smad2 Increase of TGF-β and PI3K-AKT-NF-κB pathway Down regulated Wang et al, 2020
V Human lung fibroblasts miRNA-155 Anti-fibrotic Smad1 Regulation of fibroblast proliferation, migration, and collagen synthesis Down regulated Chi et al., 2019
VI Human HEK-293T and pulmonary fibroblast line miRNA-27a-3p Anti-fibrotic Smad2 and Smad4 Regulation of myofibroblast differentiation and expression of type 1 collagen, fibronectin and α -SMA Down regulated Cui et al., 2016
VII Bleomycin murine model miRNA-19a- 19b-20a sub-cluster Anti-fibrotic TGF-βRI and TGF-βRII Regulation of pro-fibrotic genes, including
COL1A1 and CTGF
Down regulated Souma et al., 2018
VIII Bleomycin murine model and NIH/3T3 cells miRNA-29b Anti-fibrotic COL1A1 and COL3A1 Collagen expression
Regulation
Down regulated Yamada et al., 2017
IX Bleomycin murine model miRNA-18a-5p Anti-fibrotic TGF-βRII EMT regulation Down regulated Zhang et al., 2017
X Bleomycin murine model miRNA-200b/c Anti-fibrotic ZEB1 and ZEB2 Regulation of E-cadherin expression and EMT Down regulated Cao et al., 2018
XI Bleomycin murine model miRNA-27b Anti-fibrotic TGFBR1 and Smad2 Regulation of fibroblasts activation and expression of COL1A1, COL3A1, COL4A1 and α- SMA Down regulated Zeng et al., 2017
XII Human lung fibrotic samples miRNA-185 and miRNA-186 Anti-fibrotic COL5A1 Regulation of collagen V expression and EMT Down regulated Lei et al., 2016
XIII Human lung fibrotic samples miRNA-130b-3p Anti-fibrotic IGF-1 Regulation of fibroblasts activation and EMT Down regulated Li et al., 2016
XIV Human lung fibrotic samples and bleomycin murine model miRNA-101 Anti-fibrotic Wnt and Smad 2/3 Regulation of fibroblasts proliferation Down regulated Huang et al., 2017
XV Human lung fibrotic tissue and bleomycine murine model miRNA-323a-3p Anti-fibrotic Smad2 Regulation of fibroblasts proliferation Down regulated Ge at al., 2016
XVI Human lung fibrotic samples and A549 cells miRNA-221 Anti-fibrotic HMGA2 EMT regulation Down regulated Wang et al., 2016
XVII Bleomycin murine model and TGF-β treated human embryonic lung fibroblasts miRNA-182-5p Pro-fibrotic Smad2 and Smad7 Increases the expression of profibrotic proteins such as fibronectin,
α-SMA, Smad2 and Smad3
Inhibition of Smad7
Up regulated Chen et al, 2019
XVIII Bleomycin murine model and human lung fibroblast miRNA-21 Pro-fibrotic ADAMTS-1 Promotes interstitial fibroblasts proliferation and increase deposition of ECM Up regulated Zhou et al., 2018
XIX Bleomycin murine model miRNA-410 Pro-fibrotic TGF-β/ ADAMTS1 pathway Increases extracellular matrix proteins deposition and fibroblasts proliferation Up regulated Liu et al., 2018
XX Human lung fibroblasts miRNA-424 Pro-fibrotic Slit2 Positive feed-back regulation of TGF-ß-induced expression of myofibroblast differentiation markers including α
-SMA and CTGF
Up regulated Huang 2020
XXI A549 cells miRNA-31 Pro-fibrotic Smad2, Smad4 and Smad6 Promotes profibrotic factors expression like MMP7 Inhibits the expression of Smad6 Up regulated Wang et al, 2019
MiRNAs involved in the EMT in a non Smad-dependent pathway
XXII TGF-β treated human lung fibroblasts miRNA-340-5p Anti-fibrotic p38/ ATF1 signaling axis Reduces extracellular matrix deposition, fibroblasts markers such as α-SMA and COL1 and reduces fibroblasts proliferation, and viability Up regulated Wei et al., 2020
XXIII Bleomycin murine model miRNA-448 Anti-fibrotic JNK Increased cellular apoptosis, decrease of cell viability and reduced collagen synthesis by fibroblasts Up regulated Xu et al., 2020
XXIV Lipopolysaccharide fibrosis murine model miRNA-506 Anti-fibrotic P65 (NF-KB subunit) Decreased fibroblasts proliferation and EMT Down regulated Zhu et al., 2019
MiRNAs involved in the EMT involved in other signaling pathways
XXV Bleomycin murine model miRNA-152–3p, miRNA-140–3p, miRNA-148 b-3p, and miRNA-7a-5p
_______
miRNA-34a-5p, miRNA-27 b-3p, miRNA-323–3p, miRNA-27a-3p, miRNA-
34c-5p, miRNA-128–3p, and miRNA-224–5p
Anti-fibrotic
_______
Anti-fibrotic
Klf4 gene
______
Ing5 gene
EMT induction by downregulation of
TGF- β and Wnt/β- catenin pathways
________
EMT induction
Through activation of PI3K/AKT and Wnt/β-
catenin pathways
Down regulated
_______
Down regulated
Wang et al., 2020
XXVI Human BAL and THP-1 cells miRNA-185 Anti-fibrotic AKT and COL1A1 activation through the down regulation of miR-29 Decreased collagen expression and extracellular matrix deposition Down regulated Tsitoura et al., 2016
XXVII Bleomycin in vitro model (MLE-12 cell line) miRNA-142-3p Anti-fibrotic PI3K/AKT/mTOR Decreased cellular apoptosis and increase of of IL-1 and TNF-α levels Down regulated Guo et al., 2017
XXVIII Mice and human lung tissue samples miRNA-29c Anti-fibrotic Foxo3a Decreased cellular apoptosis Down regulated Xie et al., 201
XXIX Bleomycin murine model miRNA-124-3p Pro fibrotic AXIN1 TGF-β1 induced fibroblasts differentiation and increased Wnt pathway Up regulated Lu et al., 2019
XXX Bleomycin murine model and human fibrotic lung sample miRNA-301 Pro-fibrotic TSC1/mTOR pathway Promotes fibroblasts activation, proliferation and trans differentiation into myofibroblasts and collagen deposition Up regulated Wang et al., 2020

miRNA: microRNA; TGFBRI: type I TGF-β receptor; TGFBRII: type II TGF-β receptor; α-SMA: alpha-smooth muscle actin ; TGF-β1: transforming growth factor beta 1; CTGF: connective tissue growth factor; PI3K-AKT-FOXO3a-mTOR: phosphatidylinositol 3-kinase, protein kinase B, forkhead box O3, mammalian target of rapamycin; NF-kb: nuclear factor kappa-light-chain-enhancer of activated B cells; ZEB 1 / 2: zinc finger E-Box binding homeobox 1 and 2; EMT: epithelial mesenchymal transition; IGF-1: insulin-like growth factor-1; Wnt: wingless and int-1; HMGA2: high-mobility group AT-hook2; ADAMTS-1: metallopeptidase with thrombospondin type motif 1; ECM: extracellular matrix; MMP: matrix metallopeptidase; JNK: c-Jun N-terminal kinase; Klf4: kruppel-like factor 4; Ing5: growth family member5; BAL: bronchoalveolar lavage; IL1: interleukin 1; TNF-α: tumor necrosis factor alpha; AXIN: axis inhibition protein 1; TSC1: tuberous sclerosis complex.

MiRNAs involved in EMT in a Smad-dependent way

There are some miRNAs that have been studied in different samples like in a bleomycin murine model, in human A549 cells, bronchial epithelial cells and mice fibroblasts respectively for their antifibrotic role in IPF due to their up regulation, such as miRNA-877-3p, miRNA-1343 and miRNA-133.

MiRNA-877-3p can inhibit TGF-β-induced myofibroblast differentiation through the up regulation of Smad717, miRNA-1343 represses both TGF-β I and II receptor thus reducing the expression of fibrosis markers such as a-SMA and COL1A18 and miRNA-133 inhibits myofibroblast differentiation through the down regulation of profibrotic genes such as COL1A1 and CTGF19.

On the other hand, there are miRNAs that even though have an antifibrotic role in health, they favor the development of IPF due to their downregulation status in this disease such as, miRNA-184, miRNA-155 miRNA-27a-3p, miRNA-19a- 19b-20a sub-cluster, miRNA-29b, miRNA-18a-5p, miRNA-200b/c, miRNA-27b, miRNA-185, miRNA-186, miRNA-130b-3p, miRNA-101, miRNA-323a-3p and miRNA-221.

MiRNA-184, studied in A549 cells, is not able to regulate Akt and Smad220, miRNA-155 studied in human lung fibroblasts, is not able to regulate Smad121 and miRNA-27a-3p studied in a pulmonary fibroblasts line can not inhibit, the phenotypic marker of myofibroblasts, a -SMA, Smad2 and Smad422. These three miRNAs generate an induction of EMT transition, fibroblast proliferation and migration and collagen synthesis.

The next miRNAs have been studied in a bleomycin murine model: MiRNA-19a- 19b-20a sub-cluster is not able to suppress pro-fibrotic genes, including COL1A1 and CTGF23, miRNA-29b is not able to suppress the expression COL1A1 and COL3A24 and miRNA-18a-5p contributes with EMT because it is not able to regulate TGF-β type II receptor which might therefore provide a novel approach to the treatment of IPF25. MiRNA-200b/c is a possible therapeutic target too because this miRNA attenuates early pulmonary fibrosis by inhibiting ZEB1/2 via Smad-mediated way with the consequent inhibition of the expression of E-cadherin, however its expression is diminished in a fibrosis status26, and MiRNA-27b role is affected because it cannot inhibit the expression of collagens genes such as COL1A1, COL3A1, COL4A1 and a-SMA through the regulation of Smad2, that is the reason why it has been proposed as a possible therapeutic target as well27.

MiRNA-185, miRNA-186 and miRNA-130b-3p have been studied on human lung fibrotic samples and it has been seen that both miRNA-185 and miRNA-186 fail on the regulation of COL5A1 gene28 and miRNA-130b-3p is unable to regulate the action of IGF-1 on EMT process29. MiRNA-101 and miRNA-323a-3p have been studied human lung fibrotic samples but also in a bleomycin murine model, miRNA-101 acts via the Wnt and TGF-β/Smad2/3 pathways but its downregulated status generates an increase in fibroblasts proliferation, collagen synthesis and elevation of a-SMA30 and miRNA-323a-3p is not able to regulate Smad2/3 complex therefore it cannot suppress fibroblast differentiation and expression of matrix proteins.31 MiRNA-221 is another one studied on human lung fibrotic samples but also in A549 cells and it was seen that it cannot regulate EMT process because it not able to regulate HMGA232.

Finally, there are some miRNAs which have a pro fibrotic role due to their up regulation such as miRNA-182-5p, miRNA-21, miRNA-410, miRNA-424 and miRNA-31.

MiRNA-182-5p and miRNA-21 have been studied in a bleomycin murine model but also in human lung fibroblasts and it was found that miRNA-182-5p has a direct effect stimulating on Smad2/3 complex and inhibiting Smad733 and miRNA-21 causes an abnormal deposition of ECM through the increase of TGF-β/ADAMTS-1 signal pathway and inhibition of Smad7 too34. MiRNA-410 was studied in a bleomycin model and is involved in the activation of TGF-β1/ADAMTS-1 signaling pathway as well and it has a promising therapeutic future because through the inhibition of this miRNA there would be a regulation of extracellular matrix proteins like COL1 and COL3 and fibroblasts proliferation35.

In some recent studies, there has been found that miRNA-424 has an up regulated status in human lung fibroblasts due to Slit2 inhibition which generates a positive feed-back regulation of the TGF-ß-induced expression of myofibroblast differentiation markers including a-SMA and CTGF36. and miRNA-31 promotes Smad2 and Smad4 heterodimers formation and inhibits Smad6 in A 549 cells20.

miRNAs involved in EMT in a non Smad-dependent way

In recent studies it has been demonstrated that there are some miRNAs that act on non-Smad dependent pathways with an antifibrotic role like miRNA-340-5p, miRNA-448 and miRNA-506.

MiRNA-340-5p which is upregulated in TGF-β treated human lung fibroblasts regulates the ATF1/p38 signaling axis that reduces extracellular matrix, fibroblasts markers such as a-SMA and COL-1and reduces fibroblasts proliferation37.

MiRNA-448 studied in mice fibrotic tissue, was found to be upregulated and its function is to suppress the JNK signaling pathway that can inhibit IPF progression by downregulating cell proliferation and collagen synthesis by promoting apoptosis in fibroblasts38.

On the other hand, miRNA-506 has an antifibrotic role through the inhibition of p65 which is a subunit of NF-kB, however its downregulated status in a fibrosis murine model promotes fibroblasts recruitment, EMT and decreases fibroblast apoptosis39.

MiRNAs involved in EMT through other signaling pathways

There are some several miRNAs that have an antifibrotic role, however, due to their down regulated status in lung fibrosis they are not able to fulfill their role.

For example, miRNA-152–3p, miRNA-140–3p, miRNA-148 b-3p, and miRNA-7a-5p studied in bleomycin murine model, are not able to downregulate Kruppel-like factor 4 (Klf4), a negative regulator of a-SMA which has a direct effect on SNAI2, therefore suppressing TGFβ1-induced EMT and they are not able to regulate the Wnt/b- catenin pathway either. Also, miRNA-34a-5p, miRNA-27 b-3p, miRNA-323–3p, miRNA-27a-3p, miRNA-34c-5p, miRNA-128–3p, and miRNA-224–5p cannot regulate the growth family member5 (Ing5) which regulates PI3K/AKT and Wnt/β-catenin pathway therefore there is an increment in the fibrosis process40.

MiRNA-185 downregulation in human bronchoalveolar lavage (BAL) contributes to collagen deposition due to AKT overactivation and miRNA-29a downregulation which causes the overexpression of COL1A1 gene, therefore this miRNA has been proposed as possible diagnosis biomarkers for patients with IPF41. Moreover, with miRNA-142-3p mimic transfection, in bleomycin-induced injuries in an in vitro IPF model it was possible to alleviate cell injury and production of pro-inflammatory factors through the regulation of PI3K/AKT, which indicated the protective role of this miRNA, however, even though it has a anti fibrotic role it is diminished in a fibrosis state42. Another example is miRNA-29c which have an impact on cell apoptosis of AECS2 because its function is to regulate FoxO3a, however, this miRNA is negatively regulated in human lung tissue resulting in a resistance to apoptosis and lack of regulation of ECM-related target genes such as COL3A43.

Finally, there are some miRNAs which have a pro fibrotic role due to their up regulation such as miRNA-124-3p and miRNA-301a, both have been studied on a bleomycin model and miRNA-301a has also been studied on human lung fibrotic tissue samples.

MiRNA-124-3p plays an important role in TGF-β1 induced fibrogenic cell differentiation through the regulation of AXIN1 protein, which is involved in the activation of Wnt pathway44 and miRNA-301a promotes the up regulation of TSC1 gene which acts through the mTOR signaling pathway promoting fibroblasts activation and has been proposed as a possible therapeutic target45.

Results

After reviewing fifty original articles about MiRNAs involved in idiopathic pulmonary fibrosis, it was decided to include only those that related miRNAs to the main signaling pathways involved in the fibrotic process of lung parenchyma, such as TGF-β, Wnt/β-catenin and PI3K-Akt-FOXO3a-mTOR pathways.

MiRNAs are short, non-coding RNAs with an extension of approximately 21-23 nucleotides which play an important role in the genesis and development of IPF thanks to their supra regulated state by favoring epithelial mesenchymal transition and fibroblasts stimulation with the consequent synthesis of extracellular matrix proteins such as collagen; however, there is another pro fibrotic mechanism in which miRNAs cannot exert their antifibrotic effect because they are in an infra regulated state, which generates an inadequate control of intracellular receptors of the signaling pathways which will favor lung fibrosis.

Among these signaling pathways there is the TGF-β pathway, which exert its function through a Smad and non Smad dependent way. It is the main pathway involved in the regulation of the expression of profibrotic genes as well as the differentiation and proliferation of fibroblasts and the consequent deposition of collagen. There are also other alternative pathways such as the Wnt/β-catenin pathway and the PI3K-Akt-FOXO3a-mTOR pathway, which are also stimulated by miRNAs, and they related to alveolar repair through fibroblasts proliferation and extracellular matrix proteins production which generates pulmonary fibrosis.

Discussion

IPF is a progressive and degrading interstitial lung disease with poor prognosis and few therapeutic options. Nowadays it has been proposed that several miRNAs play an important role in the pathogenesis of this disease, both in human and murine fibrosis models, because they are able to regulate cellular processes such as cellular apoptosis, proliferation, and differentiation through the stimulation of different receptors within signaling pathways which causes a remodeling effect of lung parenchyma.

In this review the current knowledge about these miRNAs was grouped and synthesized to have a better understanding about their different mechanisms of action within the main signaling pathways which have as common end the trans differentiation of fibroblasts into myofibroblasts and the synthesis of extracellular matrix proteins that contribute with the fibrotic process.

Conclusions

MiRNAs have been studied in IPF, to expand the knowledge about the pathogenesis of this disease but also, they have been postulated as a new therapeutic option because nowadays the treatment of this disease is not very effective and is not curative either, so if it is possible to modify or block the action of a specific miRNA it could generate a change in the mechanism of the disease. Although it is not yet known whether this therapy will have adverse effects, it is believed that therapy based on miRNAs could be the basis of an effective treatment for pulmonary fibrosis since there is a precedent regarding the possible utility of miRNAs as therapeutic agents. This has been studied in different types of cancer where in which it is used miRNA mimic therapy and viral vectors containing the miRNA target to restore the down regulated status of anti fibrotic miRNAs and anti-miRNAs to block the pro fibrotic role of supra regulated miRNAs. However, it is suggested to expand the studies about miRNAs involved in IPF to continue updating information about different mechanisms of action altered in this pathology so that in the future, being able to help patients suffering from this chronic and devasting disease by proposing a new therapeutic agent more effective than those existing today and to provide a better quality of life.

Acknowledgements

The INER is thanked for being the source of inspiration for this article.

REFERENCES

1. Selman M, Pardo A. The leading role of epithelial cells in the pathogenesis of idiopathic pulmonary fibrosis. Cell Signal. 2020;66:109482 [ Links ]

2. Nemeth J, Schundner A, Quast K, Winkelmann VE and Frick M. A Novel Fibroblast Reporter Cell Line for in vitro Studies of Pulmonary Fibrosis. Front. Physiol. 2020;11:567675 [ Links ]

3. Calin GA, Sevignani C, Dumitru CD, Hyslop T, Noch E, Yendamuri S, et al. Human microRNA genes are frequently located at fragile sites and genomic regions involved in cancers. Proc Natl Acad Sci USA. 2004;101(9):2999-3004 [ Links ]

4. Thiery JP, Acloque H, Huang RY, Nieto MA. Epithelial-mesenchymal transitions in development and disease. Cell. 2009;139(5):871-890 [ Links ]

5. Nieto MA, Huang RY, Jackson RA, Thiery JP. EMT. Cell. 2016;166(1):21-45 [ Links ]

6. Pandit KV, Milosevic J. MicroRNA regulatory networks in idiopathic pulmonary fibrosis. Biochem Cell Biol. 2015;93(2):129-137 [ Links ]

7. Bourd-Boittin K, Bonnier D, Leyme A, Mari B, Tuffery P, Samson M, et al. Protease profiling of liver fibrosis reveals the ADAM metallopeptidase with thrombospondin type 1 motif, 1 as a central activator of transforming growth factor beta. Hepatology. 2011;54(6):2173-84. [ Links ]

8. Rajasekaran S, Rajaguru P, Sudhakar Gandhi PS. MicroRNAs as potential targets for progressive pulmonary fibrosis. Front. Pharmacol. 2015;6:254 [ Links ]

9. Chanda D, Otoupalova E, Smith SR, Volckaert T, De Langhe SP, Thannickal VJ. Developmental pathways in the pathogenesis of lung fibrosis. Mol Aspects Med. 2019;65:56-69. [ Links ]

10. Kang H. Role of MicroRNAs in TGF-b Signaling Pathway-Mediated Pulmonary Fibrosis. Int J Mol Sci. 2017;18(12):2527. [ Links ]

11. Lifshitz V, Frenkel D. Handbook of Biologically Active Peptides. Academic Press. Loa Angeles., United States of America, 2013 [ Links ]

12. Derynck, R, Zhang Y. Smad-dependent and Smad-independent pathways in TGF-b family signalling. Nature 2003;425:577–584 [ Links ]

13. Skronska-Wasek W, Gosens R, Königshoff M, Baarsma HA. WNT receptor signalling in lung physiology and pathology. Pharmacol Ther. 2018;187:150-166 [ Links ]

14. Königshoff, M, Balsara, N, Pfaff, EM, Kramer M, Chrobak, I, Seeger W, et al. Functional Wnt signaling is increased in idiopathic pulmonary fibrosis. PloS one. 2008;3(5):e2142 [ Links ]

15. Lu Y, Azad N, Wang L, Iyer AK, Castranova V, Jiang BH, et al. Phosphatidylinositol-3-kinase/akt regulates bleomycin-induced fibroblast proliferation and collagen production. Am J Respir Cell Mol Biol. 2010;42:432–441 [ Links ]

16. Laplante M, Sabatini DM. mTOR signaling in growth control and disease. Cell. 2012;149(2):274-293 [ Links ]

17. Wang C, Gu S, Cao H, Li Z, Xiang Z, Hu, K, et al. miR-877-3p targets Smad7 and is associated with myofibroblast differentiation and bleomycin-induced lung fibrosis. Sci Rep. 2016;6:30122 [ Links ]

18. Stolzenburg LR, Wachtel S, Dang H, Harris A. miR-1343 attenuates pathways of fibrosis by targeting the TGF-b receptors. Biochem J. 2016;473(3):245-256 [ Links ]

19. Wei P, Xie Y, Abel PW, Huang Y, Ma Q, Li L, et al. Transforming growth factor (TGF)-b1-induced miR-133a inhibits myofibroblast differentiation and pulmonary fibrosis. Cell Death Dis. 2019;10(9):670 [ Links ]

20. Wang CJ, Li BB, Tan YJ, Zhang GM, Cheng GL, Ren YS. MicroRNA-31/184 is involved in transforming growth factor-b-induced apoptosis in A549 human alveolar adenocarcinoma cells. Life Sci. 2020;242:117205 [ Links ]

21. Chi L, Xiao Y, Zhu L, Zhang M, Xu B, Xia H, et al. microRNA-155 attenuates profibrotic effects of transforming growth factor-beta on human lung fibroblasts. J Biol Regul Homeost Agents. 2019;33(5):1415-1424 [ Links ]

22. Cui H, Banerjee S, Xie N, Ge J, Liu R.M, Matalon S, et al. MicroRNA-27a-3p Is a Negative Regulator of Lung Fibrosis by Targeting Myofibroblast Differentiation. Am J Respir Cell Mol Biol. 2016;54(6):843-852 [ Links ]

23. Souma K, Shichino S, Hashimoto S, Ueha S, Tsukui T, Nakajima T, et al. Lung fibroblasts express a miR-19a-19b-20a sub-cluster to suppress TGF-b-associated fibroblast activation in murine pulmonary fibrosis. Sci Rep. 2018;8(1):16642 [ Links ]

24. Yamada Y, Takanashi M, Sudo K, Ueda S, Ohno SI, Kuroda M, et al. Novel form of miR-29b suppresses bleomycin-induced pulmonary fibrosis. PLoS One. 2017;12(2):e0171957 [ Links ]

25. Zhang Q, Ye H, Xiang F, Song LJ, Zhou LL, Cai PC, et al. miR-18a-5p Inhibits Sub-pleural Pulmonary Fibrosis by Targeting TGF-b Receptor II. Mol Ther. 2017;25(3):728-738. [ Links ]

26. Cao Y, Liu Y, Ping F, Yi L, Zeng Z, Li Y, et al. miR-200b/c attenuates lipopolysaccharide-induced early pulmonary fibrosis by targeting ZEB1/2 via p38 MAPK and TGF-b/smad3 signaling pathways. Lab Invest. 2018;98(3):339-359 [ Links ]

27. Zeng X, Huang C, Senavirathna L, Wang P, Liu L, et al. miR-27b inhibits fibroblast activation via targeting TGFb signaling pathway. BMC Cell Biol. 2017;18(1):9 [ Links ]

28. Lei GS, Kline HL, Lee CH, Wilkes DS, Zhang C, et al. MiR-185 and miR-186 during Idiopathic Pulmonary Fibrosis. Am J Pathol. 2016;186(9):2310-2316 [ Links ]

29. Li S, Geng J, Xu X, Huang X, Leng D, Jiang D, et al. MiR-130b-3p Modulates Epithelial-Mesenchymal Crosstalk in Lung Fibrosis by Targeting IGF-1. PLoS One. 2016;11(3):e0150418. [ Links ]

30. Huang C, Xiao X, Yang Y, Mishra A, Liang Y, Zeng X, et al. MicroRNA-101 attenuates pulmonary fibrosis by inhibiting fibroblast proliferation and activation. J Biol Chem. 2019 Apr 26;294(17):6694]. J Biol Chem. 2017;292(40):16420-16439 [ Links ]

31. Ge L, Habiel DM, Hansbro PM, Kim RY, Gharib SA, Edelman JD, et al. MiR-323a-3p regulates lung fibrosis by targeting multiple profibrotic pathways. JCI Insight. 2016;1(20):e90301 [ Links ]

32. Wang YC, Liu JS, Tang HK, Nie J, Zhu JX, Wen LL, et al. miR221 targets HMGA2 to inhibit bleomycininduced pulmonary fibrosis by regulating TGFb1/Smad3-induced EMT. Int J Mol Med. 2016;38(4):1208-1216 [ Links ]

33. Chen Y, Zhang Q, Zhou Y, Yang Z, Tan M. Inhibition of miR-182-5p attenuates pulmonary fibrosis via TGF-b/Smad pathway. Hum Exp Toxicol. 2020;39(5):683-695 [ Links ]

34. Zhou J, Xu Q, Zhang Q, Wang Z, Guan S. A novel molecular mechanism of microRNA-21 inducing pulmonary fibrosis and human pulmonary fibroblast extracellular matrix through transforming growth factor b1-mediated SMADs activation. J Cell Biochem. 2018;119(9):7834-7843 [ Links ]

35. Liu H, He Y, Jiang Z, Shen S, Mei J, Tang M. Prodigiosin Alleviates Pulmonary Fibrosis Through Inhibiting miRNA-410 and TGF-b1/ADAMTS-1 Signaling Pathway. Cell Physiol Biochem. 2018;49(2):501-511 [ Links ]

36. Huang Y, Xie Y, Abel PW, Wei P, Plowman J, Toews ML, et al. TGF-b1-induced miR-424 promotes pulmonary myofibroblast differentiation by targeting Slit2 protein expression. Biochem Pharmacol. 2020;180:114172. [ Links ]

37. Wei YQ, Guo YF, Yang SM, Ma HH, Li J. MiR-340-5p mitigates the proliferation and activation of fibroblast in lung fibrosis by targeting TGF-b/p38/ATF1 signaling pathway. Eur Rev Med Pharmacol Sci. 2020;24(11):6252-6261 [ Links ]

38. Xu F, Xu F, Xie S, Zuo W, Wen G, Zhao T, et al. MicroRNA-448 overexpression inhibits fibroblast proliferation and collagen synthesis and promotes cell apoptosis via targeting ABCC3 through the JNK signaling pathway. J Cell Physiol. 2020;235(2):1374-1385 [ Links ]

39. Zhu M, An Y, Zhang X, Wang Z, Duan H. Experimental pulmonary fibrosis was suppressed by microRNA-506 through NF-kappa-mediated apoptosis and inflammation. Cell Tissue Res. 2019;378(2):255-265 [ Links ]

40. Wang C, Cao H, Gu S, Shi C, Chen X, Han X. Expression analysis of microRNAs and mRNAs in myofibroblast differentiation of lung resident mesenchymal stem cells. Differentiation. 2020;112:10-16 [ Links ]

41. Tsitoura E, Wells AU, Karagiannis K, Lasithiotaki I, Vasarmidi E, Bibaki E, et al. MiR-185/AKT and miR-29a/collagen 1a pathways are activated in IPF BAL cells. Oncotarget. 2016;7(46):74569-74581 [ Links ]

42. Guo F, Lin SC, Zhao MS, Yu B, Li XY, Gao Q, et al. microRNA-142-3p inhibits apoptosis and inflammation induced by bleomycin through down-regulation of Cox-2 in MLE-12 cells. Braz J Med Biol Res. 2017;50(7):e5974 [ Links ]

43. Xie T, Liang J, Geng Y, Liu N, Kurkciyan A, Kulur V, et al. MicroRNA-29c Prevents Pulmonary Fibrosis by Regulating Epithelial Cell Renewal and Apoptosis. Am J Respir Cell Mol Biol. 2017;57(6):721-732 [ Links ]

44. Lu Y, Zhang T, Shan S, Wang S, Bian W, Ren T, et al. MiR-124 regulates transforming growth factor-b1 induced differentiation of lung resident mesenchymal stem cells to myofibroblast by repressing Wnt/b-catenin signaling. Dev Biol. 2019;449(2):115-121 [ Links ]

45. Wang J, Li X, Zhong M, Wang Y, Zou L, Wang M, et al. MiR-301a Suppression within Fibroblasts Limits the Progression of Fibrosis through the TSC1/mTOR Pathway. Mol Ther Nucleic Acids. 2020;21:217-228 [ Links ]

FundingThere was no funding for this article.

Ethical disclosures

Protection of human and animal subjects. The authors declare that no experiments were performed on humans or animals for this study.

Confidentiality of data. The authors declare that no patient data appear in this article.

Right to privacy and informed consent. The authors declare that no patient data appear in this article.

Received: June 29, 2021; Accepted: September 30, 2021

* Correspondence: Ana R. Cadena-Suárez E-mail: anah.cdn@gmail.com

Conflict of interests

The authors declare that they have no conflict of interest.

Creative Commons License Sociedad Médica del Hospital General de Mexico. Published by Permanyer. This is an open access article under the CC BY-NC-ND license