Open access peer-reviewed chapter

The Role of miRNAs in Idiopathic Pulmonary Fibrosis

Written By

Koichi Takagi, Munekazu Yamakuchi, Teruto Hashiguchi and Hiromasa Inoue

Submitted: 20 July 2018 Reviewed: 28 November 2018 Published: 30 April 2019

DOI: 10.5772/intechopen.82771

From the Edited Volume

Interstitial Lung Diseases

Edited by Jelena Stojšić

Chapter metrics overview

1,135 Chapter Downloads

View Full Metrics

Abstract

Idiopathic pulmonary fibrosis (IPF) is a genetic heterogeneous disease with high mortality and poor prognosis. IPF is characterized by persistent fibroblasts and relentless accumulation of collagen matrix. Epithelial-mesenchymal transition (EMT) and endothelial-to-mesenchymal transition (EndoMT) contribute to the progression of the fibrotic process. There are some therapeutic drugs that delay this progress, but eradicative medicine does not exist yet. MicroRNAs (miRNAs) are short single-stranded RNAs that regulate posttranscriptional silencing. Recent reports have shown that miRNAs play important roles in the development of IPF, as different expression levels of miRNAs in blood and lung tissue from IPF patients were closely associated with the occurrence of IPF disease. In this chapter, we will discuss the role of miRNAs in the pathogenesis, diagnosis, and treatment of IPF. In particular, we will focus on the regulation of EMT/EndoMT by miRNAs.

Keywords

  • IPF
  • miRNA
  • EndoMT
  • EMT

1. Introduction

1.1 Overview of idiopathic pulmonary fibrosis

Idiopathic pulmonary fibrosis (IPF) is a genetic heterogeneous disease with high mortality and poor prognosis that is characterized by progressive scarring of the pulmonary parenchyma, which leads to progressive loss of lung function with dyspnea and hypoxemia, and ultimately respiratory failure and death. IPF is a specific form of chronic, progressive, fibrosing interstitial pneumonia of unknown cause. Disease pathogenesis involves airway tissue remodeling through the excess deposition of extracellular matrix (ECM) proteins and the formation of fibroblastic foci, evident by histology [1]. These events are occurred mainly by continuous damage to the lung epithelium, which destroys the basement membrane and activates myofibroblasts. Myofibroblasts may not only come from resident or circulating fibroblasts but also be brought by epithelial cells transdifferentiated through epithelial-mesenchymal transition (EMT) [2]. Signaling through the TGF-β pathway is thought to underlie many of these changes [3].

According to the 2018 joint statement by the American Thoracic Society (ATS), European Respiratory Society (ERS), Latin America Thoracic Association (ALAT), and Japanese Respiratory Society (JRS) [4], the diagnosis of IPF can be secured by exclusion of other known causes of interstitial lung disease (ILD) and either the presence of high resolution computed tomography (HRCT) pattern of usual interstitial pneumonia (UIP) or specific combinations of HRCT patterns and histopathology patterns in patients subjected to lung tissue sampling. Any plausible cause of secondary interstitial involvement should be excluded by a thorough medical history and other procedures such as laboratory tests or bronchoalveolar lavage (BAL) when necessary. If a specific diagnosis is not made or no potential cause for ILD is identified, then clinical findings and HRCT are considered during multidisciplinary discussion among different experts (including clinicians, radiologists, pathologists, rheumatologists, and thoracic surgeons in selected cases) [4].

1.2 Overview of miRNAs

MicroRNAs (miRNAs) are short single-stranded RNA molecules of 19–25 nucleotides in length that mediate posttranscriptional gene silencing of target genes [5]. MiRNAs have been recognized as a distinct class of small regulatory RNAs in multiple species that regulate a wide variety of functions such as cell proliferation, differentiation, apoptosis, stress response, and immune response [6, 7, 8]. MiRNAs regulate the onset and progress of a variety of diseases, such as cancer, infections, and atherosclerosis [9, 10, 11]. In lung diseases, many miRNAs contribute to the pathogenesis. For instance, miR-424/503 regulates the progression of pulmonary hypertension (PH) [12, 13]. miR-424/503 expression is reduced in the pulmonary artery of PH patients [13]. One of the targets of miR-424/503 is Rictor, and inhibition of miR-424/503 expression increased the expression of Rictor, leading to endothelial-to-mesenchymal transition (EndoMT) and migration of pulmonary artery endothelial cells [12]. In allergic asthma, miR-21, miR-126, and miR-145 are upregulated in multiple experimental asthma models [14, 15, 16], and miR-221 and miR-485-3p are upregulated in the peripheral blood of pediatric patients with asthma, compared with controls [17].

Advertisement

2. IPF and miRNAs

Profiling of miRNAs in the lung tissue from IPF patients was performed by different groups [18, 19, 20, 21, 22]. In IPF lungs, the expression of miR-21 and miR-155 was increased, whereas the expression of let-7a, miR-29, miR-30, and miR-101 was decreased [23, 24, 25]. MiR-21 expression was increased in the serum of IPF patients, and its levels correlated with a decrease in lung function [26]. In the miRNA expression profiling of sputum-derived exosomes from IPF patients, 21 miRNAs were differentially expressed, among which seven (e.g., miR-142-3p and miR-33a-5p) were upregulated and 14 (e.g., Let-7d-5p) were downregulated [27]. In bronchoalveolar lavage fluid (BALF), miR-29a and miR-185 are downregulated in IPF patients [28, 29]. However, little is known about miRNA functions in IPF; much of the functional data has been inferred from chemically induced mouse IPF models. In the lungs of mouse IPF models, miR-21, miR-9, and miR-155 were increased [19, 21, 30], and miR-26, miR-101, miR-139, miR-200, miR-326, miR-489, miR-503, and miR-708 were decreased [24, 31, 32, 33, 34, 35, 36, 37]. Furthermore, miR-21 expression was increased in myofibroblasts from IPF lungs, and its inhibition in mouse IPF models attenuated the disease severity [19]. The expression of miR-200 family members was reduced in mouse IPF models and restoration of miR-200c inhibited fibrosis [33]. Similarly, miR-26a was decreased in mouse IPF models and its inhibition caused pulmonary fibrosis, while overexpression repressed fibrotic disease [38]. Finally, miR-326 was decreased in mouse IPF models, and its administration mimicked inhibited TGF-β expression and attenuated fibrosis [34].

Advertisement

3. The role of miRNAs in IPF-contributing cells

Epithelial cells communicate each other via adhesion molecules under physiological condition. Among many adhesion molecules, E-cadherin plays an important role in maintaining tight junctions of epithelial cells. Recurrent and nonresolving injury to lung epithelial cells appears to be a key driver of pulmonary fibrosis. Recent studies have supported the concept that altered alveolar epithelial cell phenotypes, including those produced by increased endoplasmic reticulum stress, predispose to further epithelial cell injury and abnormal repair, facilitating the development of pulmonary fibrosis [39, 40], and thus suggested that the epithelial abnormalities/dysfunction underlying fibrosis should be referred to as reprogramming. The ability of epithelial cells to change into mesenchymal cells through EMT plays an important role in the development of IPF.

Myofibroblasts and related mesenchymal cells are generally accepted as the cells predominantly responsible for fibrotic destruction/distortion of the lung in IPF [41]. Proliferation of fibroblasts and their differentiation into myofibroblasts are important in the development of IPF. The stage of mesenchymal cell differentiation at which IPF myofibroblasts acquire hallmark pathological properties, the mechanisms underlying their pathological differentiation, and the roles of signals from epithelial cells, immune cells, and the matrix in this process has not been fully elucidated and requires further research.

The lung extracellular matrix (ECM) is comprised of collagens, elastin, glycoproteins, and proteoglycans, which serve as structural scaffolding for cells and provide the mechanical stability and elastic recoil necessary for proper lung function. In normal wound healing, myofibroblasts that produce ECM proteins are recruited during the active phase of repair, but are quickly removed via apoptosis once the repair process is underway. Most of the scar matrix subsequently resolves and is replaced by a more permanent tissue. In IPF, myofibroblasts are relatively resistant to apoptosis, and their persistence leads to excessive scarring [42]. A hallmark of IPF is the accumulation of myofibroblasts in clusters called fibroblastic foci and extensive ECM deposition within the interstitium, resulting in destruction of alveolar architecture.

Vascular muscularization driven by repeated small injury is the main pathogenesis of PH, leading to the sustained fibrotic process of intima [43]. Different cellular processes have been described during pulmonary artery remodeling of PH-associated IPF, including endothelial dysfunction [44], EndoMT as a source of myofibroblasts [45], and pulmonary artery smooth muscle proliferation [46].

Monocytes and macrophages have a key role in regulating tissue repair and fibrosis, and several molecular pathways regulating their activity are being investigated as potential treatments for IPF. Macrophages possess many functions that would be expected to promote fibrosis, including fibroblast proliferation [47], regulation of ECM components [48], and secretion of profibrotic cytokines and growth factors. However, macrophages may also possess antifibrotic properties [47, 49]. Several studies have attributed these opposing functions to different macrophage populations distinguished along the lines of classical (M1) and alternative (M2) macrophage activation [50, 51, 52], with M1 macrophages being antifibrotic and M2 macrophages being either profibrotic or regulatory.

Adaptive immune responses are involved in the pathological process in IPF patients. Infiltration of activated CD4+ T-cells into IPF lung is often observed before symptoms are developed [53]. Mature dendritic cells also infiltrate in the pulmonary parenchyma of IPF patients [54]. FoxP3 positive regulatory T-cells are diminished in both the circulation and BALF of IPF patients [55].

3.1 Alveolar epithelial cells

miRNA microarray analysis of lung tissue from healthy controls and IPF patients showed that the expression of several let-7 family members was lower in IPF patients [18]. The human let-7 family includes 12 members (let-7-a1, a2, a3, b, c, d, e, f1, f2, g, 1, and miR-98), located on eight different homologous chromosomes [56]. In vitro inhibition of let-7d induced an increase in mesenchymal markers in lung epithelial cell lines. The High Mobility Group A2 (HMGA2) 3′ UTR has seven conserved sites complementary to the let-7 miRNA [57], and HMGA2 was significantly overexpressed in IPF lungs compared with controls [58]. In lung epithelial cell lines, let-7d regulated HMGA2 expression, and the increase in HMGA2 after TGF-β stimulation depended on inhibition of let-7 [18]. HMGA2 facilitates the transcription of SNAI1 and TWIST, the transcriptional repressors of adherens, which are tight and desmosomal junction components, leading to reduced intracellular adhesion. Moreover, it is a mediator of TGF-β-induced EMT. Downregulation of let-7d also led to upregulation of other fibrosis-relevant targets, such as RAS, insulin-like growth factor-1 (IGF-1), and IGF-1 receptor, and thus to the profound and sustained changes in cellular phenotype that are indeed observed in IPF [18] (Figure 1).

Figure 1.

The role of miRNAs in alveolar epithelial cells for IPF.

The expression of miR-26a was significantly decreased in the lungs of mice following the administration of bleomycin [59]. Alignment of miR-26a with the HMGA2 3′UTR sequence revealed one potential conserved seed site. Transient introduction of miR-26a into A549 cells, a pulmonary epithelial cell line, led to a significant decrease in HMGA2. However, the reduction in miR-26a expression induced an obvious EMT phenotype in A549 cells together with a clear alteration in certain related proteins, including E-cadherin, vimentin, and α-SMA [59]. Loss of function of miR-26a facilitated lung epithelial cells to transform into myofibroblasts and induce pulmonary fibrosis in mice. Lin28D is also one of the conserved putative target genes of miR-26a. Inhibition of miR-26a induced Lin28D expression and caused an obvious EMT phenotype in A549 cells [31].

The miR-200 family has been suggested to regulate the progression of pulmonary fibrosis by suppressing the EMT of alveolar epithelial cells (AECs). MiR-200 family members correlated with E-cadherin expression in epithelial cells [60, 61] and negatively correlated with ZEB family expression, which has been implicated in EMT [62]. Furthermore, the miR-200 family members have been identified as EMT suppressors through direct targeting of ZEB1 and ZEB2 [63]. MiR-200 is downregulated in the lungs of IPF patients and mouse IPF models [33]. In AECs, overexpression of miR-200, particularly miR-200b and miR-200c, attenuated TGF-β1-induced mesenchymal cell morphological characteristics and TGF-β1-repressed E-cadherin expression [33].

In fibrotic lungs, TGF-β1 is overexpressed in a broad range of cells, including epithelial cells, leading to the dysregulation of normal lung homeostasis by increasing the synthesis and altering the balance of matrix metalloproteinases and their inhibitors [64]. MiR-326 targets the TGF-β1 3′UTR, and miR-326 levels inversely correlate with TGF-β1 protein levels in multiple human cell lines and in the lungs of mouse IPF models [34]. Restoration of miR-326 levels by intranasal delivery of miR-326 mimics was sufficient to inhibit TGF-β1 expression and attenuate the fibrotic response in the lungs of mouse IPF models [34]. Furthermore, miR-1343 targets the 3′UTRs of TGFBR1 and TGFBR2 and attenuates EMT and fibrogenesis by inhibiting the TGF-β1 pathway in alveolar epithelial cell lines [65].

3.2 Fibroblasts and myofibroblasts

Several studies have shown that some miRNAs including miR-21, miR-29, miR-101, miR-153, miR-154, miR-155, and miR-210 regulate the function of fibroblasts in the lungs (Figure 2).

Figure 2.

The role of miRNAs in fibroblasts for IPF.

The expression of miR-21 is increased in the lungs of mouse IPF models and of patients with IPF [19]. In mouse IPF models, miR-21 inhibition by lentiviruses encoding anti-miR-21 suppressed morphological markers of pulmonary fibrosis compared with controls [23]. TGF-β1 is the most important profibrogenic cytokine, which increases the expression of miR-21 through SMAD2/3/4 in lung fibroblasts [19] [66]. MiR-21 inhibition inversely regulates TGF-β1-induced ECM protein expression in human pulmonary fibroblast cell lines [23]. Smad7, which inhibits the proliferation of interstitial fibroblasts, is a direct target of miR-21 and negatively correlates with miR-21 in pulmonary fibroblasts [19].

Members of the miR-29 family, which consists of three mature members, miR-29a, miR-29b, and miR-29c, are among the most downregulated miRNAs in IPF lungs [66]. The expression of miR-29c is also decreased in the lungs of mouse IPF models [67]. MiR-29 has many target genes involved in the composition and regulation of ECM, thus downregulation of miR-29 is capable of contributing to excessive ECM deposition [20, 68]. In human fetal lung fibroblasts, miR-29 suppression upregulated a series of genes, such as TGF-β1 regulated genes and adhesion molecules associated with the fibrotic phenotype [18].

MiR-101 is also one of the most downregulated miRNAs in fibrotic lungs from IPF patients and mouse IPF models [24]. Hypoxia-inducible factors (HIFs) contribute to the pathogenesis of pulmonary fibrosis through Nuclear Factor-Activated T-cell (NFAT) c2 [69]. MiR-101 suppressed WNT5a-induced proliferation of lung fibroblasts via inhibition of NFATc2 signaling by targeting Frizzled receptor 4/6 and attenuated bleomycin-induced pulmonary fibrosis in mouse IPF models [24].

MiR-153 is an intragenic miRNA that is embedded in genes encoding islet-associated protein (IA)-2 and 2β. It has been shown to participate in the pathogenesis of neurodegenerative diseases [70] and inhibit the proliferation and invasion of many types of malignant cells [71]. Furthermore, TGF-β1 downregulates miR-153 expression in lung fibrosis, and miR-153 was downregulated in the fibrotic phase, which was characterized by excessive proliferation of fibroblasts and deposition of ECM in mouse IPF models [72]. However, miR-153 reduced the contractile and migratory activities of fibroblasts. TGFBR2, a transmembrane serine/threonine kinase receptor for TGF-β, has been identified as a direct target of miR-153; moreover, miR-375 regulates SMAD2/3 phosphorylation and the fibrogenic activity of lung fibroblasts through TGFBR2 inhibition [72].

The majority of increased miRNAs in IPF localizes to chromosome 14q32 and is enriched with members of the miR-154 family. TGF-β1 regulates the expression of miRNAs in the 14q32 cluster and induces the expression changes similar to those seen in IPF. TGF-β1 induces cell proliferation through CDK4 and CDKN2B, a CDK4 inhibitor, which is a miR-154 target. Furthermore, TGF-β1-induced increases in proliferation were significantly reduced in the presence of miR-154 inhibitor in fibroblasts, and miR-154-induced changes in proliferation were mediated through the activation of the WNT/β-catenin pathway in fibroblasts [73].

MiR-155 is required for normal immune function [74]. Its overexpression is associated with inflammation, autoimmunity, and cancer [75], whereas miR-155-deficient mice develop age-related airway fibrosis [76]. Liver X receptor alpha (LXRα), which is an oxysterol-activated transcription factor, has a conserved 3′UTR seed-region sequence complementary to miR-155, and inhibition of LXRα ameliorated lung fibrosis in miR-155-deficient mice [77]. However, enforced expression of miR-155 reduced the profibrotic phenotype of IPF and miR-155-deficient fibroblasts [77]. Thus, miR-155 attenuates lung fibrosis through inhibition of LXRα in lung fibroblasts.

The expression of miR-210 has been found to be increased in patients with rapidly progressive IPF, who typically have experienced hypoxia [22]. Consistently, miR-210 was upregulated in IPF fibroblasts in response to hypoxia, and its knockdown suppressed the hypoxia-induced proliferation of IPF fibroblasts. Furthermore, HIF-2α regulated miR-210 expression and miR-210-mediated proliferation of IPF fibroblasts in response to hypoxia. Finally, miR-210 promoted IPF fibroblast proliferation through repression of its downstream target, MNT [78].

3.3 Endothelial cells

PH is frequently seen in IPF patients and is commonly attributed to hypoxic vasoconstriction and capillary destruction. Pathology findings include endothelial proliferation and medial hypertrophy that exceed those expected in the setting of hypoxia [79] (Figure 3). HIF-1 activates the transcription of genes encoding angiogenic growth factors, including vascular endothelial growth factor (VEGF), angiopoietin (ANGPT), placental growth factor (PGF), and platelet-derived growth factor (PDGF), which are secreted by hypoxic cells and stimulate endothelial cells, leading to angiogenesis. Furthermore, HIF-1 mediates hypoxia-induced phenotypic changes typical of EndoMT in endothelial cells [80].

Figure 3.

The role of miRNAs in pulmonary endothelial cells for IPF.

ANGPT1 is a ligand for receptor tyrosine kinase Tie2 [81], which is expressed in endothelial cells [82]. ANGPT1-Tie2 signaling has been shown to be involved mainly in angiogenic activity and promoting maturation of blood vessels, and it is regulated by Akt and MAPK signaling [83, 84]. The activation of ANGPT1-Tie2 signaling correlates with the severity of PH [85]. Mir-34a is downregulated in rat pulmonary arterial hypertension models [86], and ANGPT1 and Tie2 are potential targets of miR-34a, as they have a conserved miR-34a seed sequence in their 3′UTR [87]. Overexpression of miR-34a also downregulated the expression of PDGFRA, one of therapeutic targets of IPF, in pulmonary artery smooth muscle cells [86].

EndoMT has been proposed to be another potential source of myofibroblasts. In EndoMT, endothelial cells lose their own markers and obtain mesenchymal phenotype. Silencing of miR-155 directly increased RhoA expression and activity in endothelial cells and affected phosphorylation of the downstream LMK. A selective Rho kinase inhibitor partly suppressed EndoMT, strengthening the notion that RhoA plays a central role in EndoMT. Overexpression of miR-155 suppressed EndoMT, hence miR-155 functions as a negative regulator of RhoA signaling in TGF-β-induced EndoMT [88]. EndoMT has been considered as a source of myofibroblasts in PH [45], and miR-155 is downregulated in IPF lungs. Thus, miR-155 has been suggested to affect PH-associated IPF through inducing EndoMT.

3.4 Inflammatory cells

Macrophages possess many functions that would be expected to promote fibrosis, including regulation of fibroblast proliferation, recruitment, and activation, direct regulation of ECM components, and secretion of profibrotic cytokines and growth factors. There are two opposite characteristic types of functional macrophages; pro-inflammatory (M1) macrophages induced by toll-like receptor (TLR) and T helper type 1 (Th1) signals and anti-inflammatory (M2) macrophages activated by Th2 signals [89] [90]. Th2 cytokines, IL-4 and IL-13 (IL-4/IL-13), contribute to extensive tissue fibrosis in mouse models [91], and IL-4/IL-13-induced macrophages activate fibroblasts by secreting pro-fibrogenic cytokines. IL-4/IL-13 induced CCL18 in lung macrophages of IPF patients, and secreted CCL18 increased the production of collagen from fibroblasts [92]. Many transcriptional factors, such as STAT (signal transducers and activator of transcription) family, regulate macrophage polarization. M1 macrophage polarization is controlled via STAT1, and M2 macrophages activation is mediated by STAT6 [93]. To maintain STAT6 phosphorylation, M2 macrophages overcome strong expression of suppressor of cytokine signaling 1 (SOCS1), resulting in the prolongation of fibrogenesis (Figure 4).

Figure 4.

The role of miRNAs in macrophages for IPF.

IL-4/IL-13 induced miR-142-5p expression and suppressed miR-130a-3p expression in macrophages [94]. Transfection of macrophages with either miR-142-5p anti-sense oligonucleotides or miR-130-3p mimics significantly reduced the expression of M2 markers induced by IL-4 and regulated macrophage pro-fibrogenesis [94]. SOCS1 3′UTR has seven conserved sites complementary to miR-142-5p RNA, and PPARγ 3′UTR has seven conserved sites complementary to miR-130a-3p RNA. The expression of SOCS1 is reduced in IPF lungs, and bleomycin-induced lung fibrosis and inflammation were enhanced in SOCS1-deficient mice compared with WT mice [95]. Thus, miR-142-5p and miR-130a-3p regulate pulmonary fibrosis through SOCS1 and PPARγ in macrophages.

Downregulation of miR-185 has been demonstrated in IPF lung tissue and alveolar macrophages [29, 96]. MiR-185 inhibited cell growth and proliferation by directly targeting AKT1 [97]. Downregulation of miR-185 resulted in AKT activation via increased DNMT1 expression, leading to promoter hypermethylation and silencing of PTEN [77]. Moreover, miR-185 directly bound and degraded the 3′UTR of AKT1 mRNA [98]. Finally, TGF-β1 inhibited the expression of miR-185 in macrophages, and thus TGF-β1 signaling has been suggested to induce proliferation and activation of macrophages through inhibition of miR-185 expression [98].

Some lymphocytic cytokines are considered profibrotic with direct effects on fibroblast and myofibroblast activity in IPF. Th1, Th2, and Th17 have been linked to IPF pathogenesis. The Th1 subset produces IL-1α, TNF-α, PDGF, and TGF-β1. The Th2 subset typical interleukins are IL-4 and IL-13, which are directly involved in fibroblast activation and ECM production. The Th17 subset indirectly promotes fibrosis by increasing TGF-β1 levels, and it is positively regulated by TGF-β1, creating a positive feedback loop [99]. In both BALF and the peripheral blood of IPF patients, the number of CD4 + CD25 + FOXP3+ regulatory T-cells (Tregs) is decreased compared with healthy people. Thus, Tregs’ numerical and functional deficiency may play a central role in the initial phases of IPF pathogenesis [55].

In CD4+ T cells, the level of miR-155 expression is positively associated with the expression of the Th2 cytokines, IL-5 and IL-13, and miR-155 suppresses inhibitors of cytokine production including SOCS1 [100]. However, Foxp3 inhibition significantly decreased miR-146a expression in Tregs. The target gene of miR-146a is TNF receptor-associated factor 6 (TRAF6). Foxp3 and miR-146a have been suggested to regulate colonic inflammation and fibrosis by negatively regulating TRAF6 [101].

Advertisement

4. Conclusions

IPF is a heterogeneous disease that is affected and developed by various genetic backgrounds and environments. The recent development and approval of antifibrotic agents such as nintedanib and pirfenidone, both of which reduced the lung function decline rate in IPF patients in clinical trials, offer hope that it may be possible to alter the increased mortality associated with IPF; however, these therapeutic effects are not sufficient for curing the disease. To reveal new therapeutic IPF targets, genetic screening of IPF patients, including miRNAs, is an effective option. Over the past decade, the role of miRNAs in both promoting and alleviating chronic respiratory disease has become increasingly apparent. In IPF, some miRNAs have been indicated to be associated with EMT, EndoMT, fibrocyte differentiation, or ECM deposition. Investigation of miRNAs and their target genes associated with IPF may lead to new therapy. However, there have been some problems with applying miRNAs in the clinical scene: (1) multiple groups performing studies with similar experimental conditions have reported different and often contrasting results. Hence, collection methods and specimen parts need to be standardized, and better assays are needed for high-confidence miRNA measurements. (2) It is not clear whether miRNA misregulation is a direct cause of disease or an indirect result of gene regulatory processes. Most studies did not attempt to establish the necessity and sufficiency of the miRNA under investigation. (3) It is still difficult to interfere with miRNA expression in the human body, and it is possible that such interference will cause unexpected adverse reactions. (4) It has not been established how to introduce miRNAs into target cells safely and effectively, and the cost of this treatment will probably be high. Thus, miRNAs are still insufficient to be used as therapeutic targets in the clinical scene. Nevertheless, miRNAs may be important tools as IPF biomarkers. To use miRNAs clinically, further studies are necessary to elucidate the association between miRNAs and IPF.

References

  1. 1. Fernandez IE, Eickelberg O. New cellular and molecular mechanisms of lung injury and fibrosis in idiopathic pulmonary fibrosis. Lancet. 2012;380(9842):680-688. DOI: 10.1016/s0140-6736(12)61144-1
  2. 2. Jin HL, Dong JC. Pathogenesis of idiopathic pulmonary fibrosis: From initial apoptosis of epithelial cells to lung remodeling. Chinese Medical Journal. 2011;124(24):4330-4338
  3. 3. Leask A, Abraham DJ. TGF-beta signaling and the fibrotic response. The FASEB Journal. 2004;18(7):816-827. DOI: 10.1096/fj.03-1273rev
  4. 4. Raghu G, Remy-Jardin M, Myers JL, Richeldi L, Ryerson CJ, Lederer DJ, et al. Diagnosis of idiopathic pulmonary fibrosis: An official ATS/ERS/JRS/ALAT clinical practice guideline. American Journal of Respiratory and Critical Care Medicine. 2018;198(5):e44-e68. DOI: 10.1164/rccm.201807-1255ST
  5. 5. Winter J, Jung S, Keller S, Gregory RI, Diederichs S. Many roads to maturity: MicroRNA biogenesis pathways and their regulation. Nature Cell Biology. 2009;11(3):228-234. DOI: 10.1038/ncb0309-228
  6. 6. Bushati N, Cohen SM. MicroRNA functions. Annual Review of Cell and Developmental Biology. 2007;23:175-205. DOI: 10.1146/annurev.cellbio.23.090506.123406
  7. 7. Leung AK, Sharp PA. MicroRNA functions in stress responses. Molecular Cell. 2010;40(2):205-215. DOI: 10.1016/j.molcel.2010.09.027
  8. 8. O'Connell RM, Rao DS, Baltimore D. MicroRNA regulation of inflammatory responses. Annual Review of Immunology. 2012;30:295-312. DOI: 10.1146/annurev-immunol-020711-075013
  9. 9. Medina PP, Nolde M, Slack FJ. OncomiR addiction in an in vivo model of microRNA-21-induced pre-B-cell lymphoma. Nature. 2010;467(7311):86-90. DOI: 10.1038/nature09284
  10. 10. Chowdhury SR, Reimer A, Sharan M, Kozjak-Pavlovic V, Eulalio A, Prusty BK, et al. Chlamydia preserves the mitochondrial network necessary for replication via microRNA-dependent inhibition of fission. The Journal of Cell Biology. 2017;216(4):1071-1089. DOI: 10.1083/jcb.201608063
  11. 11. Li XX, Liu YM, Li YJ, Xie N, Yan YF, Chi YL, et al. High glucose concentration induces endothelial cell proliferation by regulating cyclin-D2-related miR-98. Journal of Cellular and Molecular Medicine. 2016;20(6):1159-1169. DOI: 10.1111/jcmm.12765
  12. 12. Takagi K, Yamakuchi M, Matsuyama T, Kondo K, Uchida A, Misono S, et al. IL-13 enhances mesenchymal transition of pulmonary artery endothelial cells via down-regulation of miR-424/503 in vitro. Cellular Signalling. 2018;42:270-280. DOI: 10.1016/j.cellsig.2017.10.019
  13. 13. Kim J, Hwangbo C, Hu X, Kang Y, Papangeli I, Mehrotra D, et al. Restoration of impaired endothelial myocyte enhancer factor 2 function rescues pulmonary arterial hypertension. Circulation. 2015;131(2):190-199. DOI: 10.1161/circulationaha.114.013339
  14. 14. Lu TX, Munitz A, Rothenberg ME. MicroRNA-21 is up-regulated in allergic airway inflammation and regulates IL-12p35 expression. Journal of Immunology. 2009;182(8):4994-5002. DOI: 10.4049/jimmunol.0803560
  15. 15. Mattes J, Collison A, Plank M, Phipps S, Foster PS. Antagonism of microRNA-126 suppresses the effector function of TH2 cells and the development of allergic airways disease. Proceedings of the National Academy of Sciences of the United States of America. 2009;106(44):18704-18709. DOI: 10.1073/pnas.0905063106
  16. 16. Collison A, Mattes J, Plank M, Foster PS. Inhibition of house dust mite-induced allergic airways disease by antagonism of microRNA-145 is comparable to glucocorticoid treatment. The Journal of Allergy and Clinical Immunology. 2011;128(1):160-7e4. DOI: 10.1016/j.jaci.2011.04.005
  17. 17. Qin HB, Xu B, Mei JJ, Li D, Liu JJ, Zhao DY, et al. Inhibition of miRNA-221 suppresses the airway inflammation in asthma. Inflammation. 2012;35(4):1595-1599. DOI: 10.1007/s10753-012-9474-1
  18. 18. Pandit KV, Corcoran D, Yousef H, Yarlagadda M, Tzouvelekis A, Gibson KF, et al. Inhibition and role of let-7d in idiopathic pulmonary fibrosis. American Journal of Respiratory and Critical Care Medicine. 2010;182(2):220-229. DOI: 10.1164/rccm.200911-1698OC
  19. 19. Liu G, Friggeri A, Yang Y, Milosevic J, Ding Q, Thannickal VJ, et al. miR-21 mediates fibrogenic activation of pulmonary fibroblasts and lung fibrosis. The Journal of Experimental Medicine. 2010;207(8):1589-1597. DOI: 10.1084/jem.20100035
  20. 20. Cushing L, Kuang PP, Qian J, Shao F, Wu J, Little F, et al. miR-29 is a major regulator of genes associated with pulmonary fibrosis. American Journal of Respiratory Cell and Molecular Biology. 2011;45(2):287-294. DOI: 10.1165/rcmb.2010-0323OC
  21. 21. Pottier N, Maurin T, Chevalier B, Puissegur MP, Lebrigand K, Robbe-Sermesant K, et al. Identification of keratinocyte growth factor as a target of microRNA-155 in lung fibroblasts: Implication in epithelial-mesenchymal interactions. PLoS One. 2009;4(8):e6718. DOI: 10.1371/journal.pone.0006718
  22. 22. Oak SR, Murray L, Herath A, Sleeman M, Anderson I, Joshi AD, et al. A micro RNA processing defect in rapidly progressing idiopathic pulmonary fibrosis. PLoS One. 2011;6(6):e21253. DOI: 10.1371/journal.pone.0021253
  23. 23. 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 beta1-mediated SMADs activation. Journal of Cellular Biochemistry. 2018;119(9):7834-7843. DOI: 10.1002/jcb.27185
  24. 24. 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. The Journal of Biological Chemistry. 2017;292(40):16420-16439. DOI: 10.1074/jbc.M117.805747
  25. 25. Zhang S, Liu H, Liu Y, Zhang J, Li H, Liu W, et al. miR-30a as potential therapeutics by targeting TET1 through regulation of Drp-1 promoter hydroxymethylation in idiopathic pulmonary fibrosis. International Journal of Molecular Sciences. 2017;18(3). DOI: 10.3390/ijms18030633
  26. 26. Li P, Zhao GQ, Chen TF, Chang JX, Wang HQ, Chen SS, et al. Serum miR-21 and miR-155 expression in idiopathic pulmonary fibrosis. The Journal of Asthma. 2013;50(9):960-964. DOI: 10.3109/02770903.2013.822080
  27. 27. Njock MS, Guiot J, Henket MA, Nivelles O, Thiry M, Dequiedt F, et al. Sputum exosomes: Promising biomarkers for idiopathic pulmonary fibrosis. Thorax. 2018. DOI: 10.1136/thoraxjnl-2018-211897
  28. 28. Bibaki E, Tsitoura E, Vasarmidi E, Margaritopoulos G, Trachalaki A, Koutoulaki C, et al. miR-185 and miR-29a are similarly expressed in the bronchoalveolar lavage cells in IPF and lung cancer but common targets DNMT1 and COL1A1 show disease specific patterns. Molecular Medicine Reports. 2018;17(5):7105-7112. DOI: 10.3892/mmr.2018.8778
  29. 29. 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. DOI: 10.18632/oncotarget.12740
  30. 30. Dai WJ, Qiu J, Sun J, Ma CL, Huang N, Jiang Y, et al. Downregulation of microRNA-9 reduces inflammatory response and fibroblast proliferation in mice with idiopathic pulmonary fibrosis through the ANO1-mediated TGF-beta-Smad3 pathway. Journal of Cellular Physiology. 2019:234(3):2552-2565. DOI: 10.1002/jcp.26961
  31. 31. Liang H, Liu S, Chen Y, Bai X, Liu L, Dong Y, et al. miR-26a suppresses EMT by disrupting the Lin28B/let-7d axis: Potential cross-talks among miRNAs in IPF. Journal of Molecular Medicine (Berlin). 2016;94(6):655-665. DOI: 10.1007/s00109-016-1381-8
  32. 32. Wang Y, Liang Y, Luo J, Nie J, Yin H, Chen Q, et al. XIST/miR-139 axis regulates bleomycin (BLM)-induced extracellular matrix (ECM) and pulmonary fibrosis through beta-catenin. Oncotarget. 2017;8(39):65359-65369. DOI: 10.18632/oncotarget.18310
  33. 33. Yang S, Banerjee S, de Freitas A, Sanders YY, Ding Q, Matalon S, et al. Participation of miR-200 in pulmonary fibrosis. The American Journal of Pathology. 2012;180(2):484-493. DOI: 10.1016/j.ajpath.2011.10.005
  34. 34. Das S, Kumar M, Negi V, Pattnaik B, Prakash YS, Agrawal A, et al. MicroRNA-326 regulates profibrotic functions of transforming growth factor-beta in pulmonary fibrosis. American Journal of Respiratory Cell and Molecular Biology. 2014;50(5):882-892. DOI: 10.1165/rcmb.2013-0195OC
  35. 35. Liu B, Li R, Zhang J, Meng C, Zhang J, Song X, et al. MicroRNA-708-3p as a potential therapeutic target via the ADAM17-GATA/STAT3 axis in idiopathic pulmonary fibrosis. Experimental and Molecular Medicine. 2018;50(3):e465. DOI: 10.1038/emm.2017.311
  36. 36. Yan W, Wu Q, Yao W, Li Y, Liu Y, Yuan J, et al. miR-503 modulates epithelial-mesenchymal transition in silica-induced pulmonary fibrosis by targeting PI3K p85 and is sponged by lncRNA MALAT1. Scientific Reports. 2017;7(1):11313. DOI: 10.1038/s41598-017-11904-8
  37. 37. Wu Q, Han L, Yan W, Ji X, Han R, Yang J, et al. miR-489 inhibits silica-induced pulmonary fibrosis by targeting MyD88 and Smad3 and is negatively regulated by lncRNA CHRF. Scientific Reports. 2016;6:30921. DOI: 10.1038/srep30921
  38. 38. Liang H, Xu C, Pan Z, Zhang Y, Xu Z, Chen Y, et al. The antifibrotic effects and mechanisms of microRNA-26a action in idiopathic pulmonary fibrosis. Molecular Therapy. 2014;22(6):1122-1133. DOI: 10.1038/mt.2014.42
  39. 39. Selman M, Pardo A. Role of epithelial cells in idiopathic pulmonary fibrosis: From innocent targets to serial killers. Proceedings of the American Thoracic Society. 2006;3(4):364-372. DOI: 10.1513/pats.200601-003TK
  40. 40. Tanjore H, Lawson WE, Blackwell TS. Endoplasmic reticulum stress as a pro-fibrotic stimulus. Biochimica et Biophysica Acta. 2013;1832(7):940-947. DOI: 10.1016/j.bbadis.2012.11.011
  41. 41. Scotton CJ, Chambers RC. Molecular targets in pulmonary fibrosis: The myofibroblast in focus. Chest. 2007;132(4):1311-1321. DOI: 10.1378/chest.06-2568
  42. 42. Bellaye PS, Kolb M. Why do patients get idiopathic pulmonary fibrosis? Current concepts in the pathogenesis of pulmonary fibrosis. BMC Medicine. 2015;13:176. DOI: 10.1186/s12916-015-0412-6
  43. 43. Heath D, Gillund TD, Kay JM, Hawkins CF. Pulmonary vascular disease in honeycomb lung. The Journal of Pathology and Bacteriology. 1968;95(2):423-430. DOI: 10.1002/path.1700950212
  44. 44. Blanco I, Ribas J, Xaubet A, Gomez FP, Roca J, Rodriguez-Roisin R, et al. Effects of inhaled nitric oxide at rest and during exercise in idiopathic pulmonary fibrosis. Journal of Applied Physiology (Bethesda, Md: 1985). 2011;110(3):638-645. DOI: 10.1152/japplphysiol.01104.2010
  45. 45. Hashimoto N, Phan SH, Imaizumi K, Matsuo M, Nakashima H, Kawabe T, et al. Endothelial-mesenchymal transition in bleomycin-induced pulmonary fibrosis. American Journal of Respiratory Cell and Molecular Biology. 2010;43(2):161-172. DOI: 10.1165/rcmb.2009-0031OC
  46. 46. Yeager ME, Frid MG, Stenmark KR. Progenitor cells in pulmonary vascular remodeling. Pulmonary Circulation. 2011;1(1):3-16. DOI: 10.4103/2045-8932.78095
  47. 47. Song E, Ouyang N, Horbelt M, Antus B, Wang M, Exton MS. Influence of alternatively and classically activated macrophages on fibrogenic activities of human fibroblasts. Cellular Immunology. 2000;204(1):19-28. DOI: 10.1006/cimm.2000.1687
  48. 48. Atabai K, Jame S, Azhar N, Kuo A, Lam M, McKleroy W, et al. Mfge8 diminishes the severity of tissue fibrosis in mice by binding and targeting collagen for uptake by macrophages. The Journal of Clinical Investigation. 2009;119(12):3713-3722. DOI: 10.1172/jci40053
  49. 49. Murray PJ, Wynn TA. Protective and pathogenic functions of macrophage subsets. Nature Reviews. Immunology. 2011;11(11):723-737. DOI: 10.1038/nri3073
  50. 50. Murray LA, Chen Q, Kramer MS, Hesson DP, Argentieri RL, Peng X, et al. TGF-beta driven lung fibrosis is macrophage dependent and blocked by Serum amyloid P. The International Journal of Biochemistry and Cell Biology. 2011;43(1):154-162. DOI: 10.1016/j.biocel.2010.10.013
  51. 51. Murray LA, Rosada R, Moreira AP, Joshi A, Kramer MS, Hesson DP, et al. Serum amyloid P therapeutically attenuates murine bleomycin-induced pulmonary fibrosis via its effects on macrophages. PLoS One. 2010;5(3):e9683. DOI: 10.1371/journal.pone.0009683
  52. 52. Gibbons MA, MacKinnon AC, Ramachandran P, Dhaliwal K, Duffin R, Phythian-Adams AT, et al. Ly6Chi monocytes direct alternatively activated profibrotic macrophage regulation of lung fibrosis. American Journal of Respiratory and Critical Care Medicine. 2011;184(5):569-581. DOI: 10.1164/rccm.201010-1719OC
  53. 53. Rosas IO, Ren P, Avila NA, Chow CK, Franks TJ, Travis WD, et al. Early interstitial lung disease in familial pulmonary fibrosis. American Journal of Respiratory and Critical Care Medicine. 2007;176(7):698-705. DOI: 10.1164/rccm.200702-254OC
  54. 54. Marchal-Somme J, Uzunhan Y, Marchand-Adam S, Kambouchner M, Valeyre D, Crestani B, et al. Dendritic cells accumulate in human fibrotic interstitial lung disease. American Journal of Respiratory and Critical Care Medicine. 2007;176(10):1007-1014. DOI: 10.1164/rccm.200609-1347OC
  55. 55. Kotsianidis I, Nakou E, Bouchliou I, Tzouvelekis A, Spanoudakis E, Steiropoulos P, et al. Global impairment of CD4+CD25+FOXP3+ regulatory T cells in idiopathic pulmonary fibrosis. American Journal of Respiratory and Critical Care Medicine. 2009;179(12):1121-1130. DOI: 10.1164/rccm.200812-1936OC
  56. 56. Jerome T, Laurie P, Louis B, Pierre C. Enjoy the silence: The story of let-7 microRNA and cancer. Current Genomics. 2007;8(4):229-233
  57. 57. Lewis BP, Burge CB, Bartel DP. Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell. 2005;120(1):15-20. DOI: 10.1016/j.cell.2004.12.035
  58. 58. Rosas IO, Richards TJ, Konishi K, Zhang Y, Gibson K, Lokshin AE, et al. MMP1 and MMP7 as potential peripheral blood biomarkers in idiopathic pulmonary fibrosis. PLoS Medicine. 2008;5(4):e93. DOI: 10.1371/journal.pmed.0050093
  59. 59. Liang H, Gu Y, Li T, Zhang Y, Huangfu L, Hu M, et al. Integrated analyses identify the involvement of microRNA-26a in epithelial-mesenchymal transition during idiopathic pulmonary fibrosis. Cell Death and Disease. 2014;5:e1238. DOI: 10.1038/cddis.2014.207
  60. 60. Christoffersen NR, Silahtaroglu A, Orom UA, Kauppinen S, Lund AH. miR-200b mediates post-transcriptional repression of ZFHX1B. RNA. 2007;13(8):1172-1178. DOI: 10.1261/rna.586807
  61. 61. Hurteau GJ, Carlson JA, Spivack SD, Brock GJ. Overexpression of the microRNA hsa-miR-200c leads to reduced expression of transcription factor 8 and increased expression of E-cadherin. Cancer Research. 2007;67(17):7972-7976. DOI: 10.1158/0008-5472.can-07-1058
  62. 62. Spaderna S, Schmalhofer O, Hlubek F, Berx G, Eger A, Merkel S, et al. A transient, EMT-linked loss of basement membranes indicates metastasis and poor survival in colorectal cancer. Gastroenterology. 2006;131(3):830-840. DOI: 10.1053/j.gastro.2006.06.016
  63. 63. Korpal M, Lee ES, Hu G, Kang Y. The miR-200 family inhibits epithelial-mesenchymal transition and cancer cell migration by direct targeting of E-cadherin transcriptional repressors ZEB1 and ZEB2. The Journal of Biological Chemistry. 2008;283(22):14910-14914. DOI: 10.1074/jbc.C800074200
  64. 64. Kisseleva T, Brenner DA. Mechanisms of fibrogenesis. Experimental Biology and Medicine (Maywood, N.J.). 2008;233(2):109-122. DOI: 10.3181/0707-mr-190
  65. 65. Stolzenburg LR, Wachtel S, Dang H, Harris A. miR-1343 attenuates pathways of fibrosis by targeting the TGF-beta receptors. The Biochemical Journal. 2016;473(3):245-256. DOI: 10.1042/bj20150821
  66. 66. Pandit KV, Milosevic J, Kaminski N. MicroRNAs in idiopathic pulmonary fibrosis. Translational Research. 2011;157(4):191-199. DOI: 10.1016/j.trsl.2011.01.012
  67. 67. Matsushima S, Ishiyama J. MicroRNA-29c regulates apoptosis sensitivity via modulation of the cell-surface death receptor, Fas, in lung fibroblasts. American Journal of Physiology. Lung Cellular and Molecular Physiology. 2016;311(6):L1050-L1L61. DOI: 10.1152/ajplung.00252.2016
  68. 68. Montgomery RL, Yu G, Latimer PA, Stack C, Robinson K, Dalby CM, et al. MicroRNA mimicry blocks pulmonary fibrosis. EMBO Molecular Medicine. 2014;6(10):1347-1356. DOI: 10.15252/emmm.201303604
  69. 69. Senavirathna LK, Huang C, Yang X, Munteanu MC, Sathiaseelan R, Xu D, et al. Hypoxia induces pulmonary fibroblast proliferation through NFAT signaling. Scientific Reports. 2018;8(1):2709. DOI: 10.1038/s41598-018-21073-x
  70. 70. Liang C, Zhu H, Xu Y, Huang L, Ma C, Deng W, et al. MicroRNA-153 negatively regulates the expression of amyloid precursor protein and amyloid precursor-like protein 2. Brain Research. 2012;1455:103-113. DOI: 10.1016/j.brainres.2011.10.051
  71. 71. Xie T, Liang J, Guo R, Liu N, Noble PW, Jiang D. Comprehensive microRNA analysis in bleomycin-induced pulmonary fibrosis identifies multiple sites of molecular regulation. Physiological Genomics. 2011;43(9):479-487. DOI: 10.1152/physiolgenomics.00222.2010
  72. 72. Liang C, Li X, Zhang L, Cui D, Quan X, Yang W. The anti-fibrotic effects of microRNA-153 by targeting TGFBR-2 in pulmonary fibrosis. Experimental and Molecular Pathology. 2015;99(2):279-285. DOI: 10.1016/j.yexmp.2015.07.011
  73. 73. Milosevic J, Pandit K, Magister M, Rabinovich E, Ellwanger DC, Yu G, et al. Profibrotic role of miR-154 in pulmonary fibrosis. American Journal of Respiratory Cell and Molecular Biology. 2012;47(6):879-887. DOI: 10.1165/rcmb.2011-0377OC
  74. 74. Kurowska-Stolarska M, Alivernini S, Ballantine LE, Asquith DL, Millar NL, Gilchrist DS, et al. MicroRNA-155 as a proinflammatory regulator in clinical and experimental arthritis. Proceedings of the National Academy of Sciences of the United States of America. 2011;108(27):11193-11198. DOI: 10.1073/pnas.1019536108
  75. 75. O'Connell RM, Rao DS, Chaudhuri AA, Boldin MP, Taganov KD, Nicoll J, et al. Sustained expression of microRNA-155 in hematopoietic stem cells causes a myeloproliferative disorder. The Journal of Experimental Medicine. 2008;205(3):585-594. DOI: 10.1084/jem.20072108
  76. 76. Rodriguez A, Vigorito E, Clare S, Warren MV, Couttet P, Soond DR, et al. Requirement of bic/microRNA-155 for normal immune function. Science. 2007;316(5824):608-611. DOI: 10.1126/science.1139253
  77. 77. Kurowska-Stolarska M, Hasoo MK, Welsh DJ, Stewart L, McIntyre D, Morton BE, et al. The role of microRNA-155/liver X receptor pathway in experimental and idiopathic pulmonary fibrosis. The Journal of Allergy and Clinical Immunology. 2017;139(6):1946-1956. DOI: 10.1016/j.jaci.2016.09.021
  78. 78. Bodempudi V, Hergert P, Smith K, Xia H, Herrera J, Peterson M, et al. miR-210 promotes IPF fibroblast proliferation in response to hypoxia. American Journal of Physiology. Lung Cellular and Molecular Physiology. 2014;307(4):L283-L294. DOI: 10.1152/ajplung.00069.2014
  79. 79. Patel NM, Lederer DJ, Borczuk AC, Kawut SM. Pulmonary hypertension in idiopathic pulmonary fibrosis. Chest. 2007;132(3):998-1006. DOI: 10.1378/chest.06-3087
  80. 80. Xu X, Tan X, Tampe B, Sanchez E, Zeisberg M, Zeisberg EM. Snail is a direct target of hypoxia-inducible factor 1alpha (HIF1alpha) in hypoxia-induced endothelial to mesenchymal transition of human coronary endothelial cells. The Journal of Biological Chemistry. 2015;290(27):16653-16664. DOI: 10.1074/jbc.M115.636944
  81. 81. Saharinen P, Kerkela K, Ekman N, Marron M, Brindle N, Lee GM, et al. Multiple angiopoietin recombinant proteins activate the Tie1 receptor tyrosine kinase and promote its interaction with Tie2. The Journal of Cell Biology. 2005;169(2):239-243. DOI: 10.1083/jcb.200411105
  82. 82. Kugathasan L, Ray JB, Deng Y, Rezaei E, Dumont DJ, Stewart DJ. The angiopietin-1-Tie2 pathway prevents rather than promotes pulmonary arterial hypertension in transgenic mice. The Journal of Experimental Medicine. 2009;206(10):2221-2234. DOI: 10.1084/jem.20090389
  83. 83. Yuan HT, Venkatesha S, Chan B, Deutsch U, Mammoto T, Sukhatme VP, et al. Activation of the orphan endothelial receptor Tie1 modifies Tie2-mediated intracellular signaling and cell survival. The FASEB Journal. 2007;21(12):3171-3183. DOI: 10.1096/fj.07-8487com
  84. 84. Liu XB, Jiang J, Gui C, Hu XY, Xiang MX, Wang JA. Angiopoietin-1 protects mesenchymal stem cells against serum deprivation and hypoxia-induced apoptosis through the PI3K/Akt pathway. Acta Pharmacologica Sinica. 2008;29(7):815-822. DOI: 10.1111/j.1745-7254.2008.00811.x
  85. 85. Humbert M, Morrell NW, Archer SL, Stenmark KR, MR ML, Lang IM, et al. Cellular and molecular pathobiology of pulmonary arterial hypertension. Journal of the American College of Cardiology. 2004;43(12 Suppl S):13S-24S. DOI: 10.1016/j.jacc.2004.02.029
  86. 86. Wang P, Xu J, Hou Z, Wang F, Song Y, Wang J, et al. miRNA-34a promotes proliferation of human pulmonary artery smooth muscle cells by targeting PDGFRA. Cell Proliferation. 2016;49(4):484-493. DOI: 10.1111/cpr.12265
  87. 87. Syed M, Das P, Pawar A, Aghai ZH, Kaskinen A, Zhuang ZW, et al. Hyperoxia causes miR-34a-mediated injury via angiopoietin-1 in neonatal lungs. Nature Communications. 2017;8(1):1173. DOI: 10.1038/s41467-017-01349-y
  88. 88. Qu Y, Zhang G, Ji Y, Zhua H, Lv C, Jiang W. Protective role of gambogic acid in experimental pulmonary fibrosis in vitro and in vivo. Phytomedicine. 2016;23(4):350-358. DOI: 10.1016/j.phymed.2016.01.011
  89. 89. Gordon S, Martinez FO. Alternative activation of macrophages: Mechanism and functions. Immunity. 2010;32(5):593-604. DOI: 10.1016/j.immuni.2010.05.007
  90. 90. Su S, Liu Q, Chen J, Chen J, Chen F, He C, et al. A positive feedback loop between mesenchymal-like cancer cells and macrophages is essential to breast cancer metastasis. Cancer Cell. 2014;25(5):605-620. DOI: 10.1016/j.ccr.2014.03.021
  91. 91. Fichtner-Feigl S, Strober W, Kawakami K, Puri RK, Kitani A. IL-13 signaling through the IL-13alpha2 receptor is involved in induction of TGF-beta1 production and fibrosis. Nature Medicine. 2006;12(1):99-106. DOI: 10.1038/nm1332
  92. 92. Prasse A, Pechkovsky DV, Toews GB, Jungraithmayr W, Kollert F, Goldmann T, et al. A vicious circle of alveolar macrophages and fibroblasts perpetuates pulmonary fibrosis via CCL18. American Journal of Respiratory and Critical Care Medicine. 2006;173(7):781-792. DOI: 10.1164/rccm.200509-1518OC
  93. 93. Sica A, Mantovani A. Macrophage plasticity and polarization: In vivo veritas. The Journal of Clinical Investigation. 2012;122(3):787-795. DOI: 10.1172/jci59643
  94. 94. Su S, Zhao Q, He C, Huang D, Liu J, Chen F, et al. miR-142-5p and miR-130a-3p are regulated by IL-4 and IL-13 and control profibrogenic macrophage program. Nature Communications. 2015;6:8523. DOI: 10.1038/ncomms9523
  95. 95. Nakashima T, Yokoyama A, Onari Y, Shoda H, Haruta Y, Hattori N, et al. Suppressor of cytokine signaling 1 inhibits pulmonary inflammation and fibrosis. The Journal of Allergy and Clinical Immunology. 2008;121(5):1269-1276. DOI: 10.1016/j.jaci.2008.02.003
  96. 96. Lei GS, Kline HL, Lee CH, Wilkes DS, Zhang C. Regulation of collagen V expression and epithelial-mesenchymal transition by miR-185 and miR-186 during idiopathic pulmonary fibrosis. The American Journal of Pathology. 2016;186(9):2310-2316. DOI: 10.1016/j.ajpath.2016.04.015
  97. 97. Qadir XV, Han C, Lu D, Zhang J, Wu T. miR-185 inhibits hepatocellular carcinoma growth by targeting the DNMT1/PTEN/Akt pathway. The American Journal of Pathology. 2014;184(8):2355-2364. DOI: 10.1016/j.ajpath.2014.05.004
  98. 98. Takahashi Y, Forrest AR, Maeno E, Hashimoto T, Daub CO, Yasuda J. miR-107 and MiR-185 can induce cell cycle arrest in human non small cell lung cancer cell lines. PLoS One. 2009;4(8):e6677. DOI: 10.1371/journal.pone.0006677
  99. 99. Betensley A, Sharif R, Karamichos D. A systematic review of the role of dysfunctional wound healing in the pathogenesis and treatment of idiopathic pulmonary fibrosis. Journal of Clinical Medicine. 2016;6(1). DOI: 10.3390/jcm6010002
  100. 100. Daniel E, Roff A, Hsu MH, Panganiban R, Lambert K, Ishmael F. Effects of allergic stimulation and glucocorticoids on miR-155 in CD4(+) T-cells. American Journal of Clinical and Experimental Immunology. 2018;7(4):57-66
  101. 101. Wang J, Yang L, Wang L, Yang Y, Wang Y. Forkhead box p3 controls progression of oral lichen planus by regulating microRNA-146a. Journal of Cellular Biochemistry. 2018;119(11):8862-8871. DOI: 10.1002/jcb.27139

Written By

Koichi Takagi, Munekazu Yamakuchi, Teruto Hashiguchi and Hiromasa Inoue

Submitted: 20 July 2018 Reviewed: 28 November 2018 Published: 30 April 2019