RESEARCH ARTICLE


Role of MicroRNAs in Fibrosis



Serena Vettori 1, 2, Steffen Gay 1, Oliver Distler*, 1
1 Center of Experimental Rheumatology, ZIHP, University Hospital Zurich, Gloriastrasse 25, CH-8091 Zurich, Switzerland
2 Department of Clinical Medicine, Cardiovascular and Immunologic Sciences, University of Naples “Federico II”, via Pansini 5, 80131 I-Naples, Italy


Article Metrics

CrossRef Citations:
130
Total Statistics:

Full-Text HTML Views: 2809
Abstract HTML Views: 2458
PDF Downloads: 717
Total Views/Downloads: 5991
Unique Statistics:

Full-Text HTML Views: 1308
Abstract HTML Views: 1432
PDF Downloads: 493
Total Views/Downloads: 3240



Creative Commons License
© Vettori et al.; Licensee Bentham Open.

open-access license: This is an open access article licensed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/3.0/) which permits unrestricted, non-commercial use, distribution and reproduction in any medium, provided the work is properly cited.

* Address correspondence to this author at the University Hospital Zurich, Gloriastrasse 25, 8091 Zurich, Switzerland; Tel: +41-44-255-2977; Fax: +41-44-255-4170; E-mail: oliver.distler@usz.ch


Abstract

Fibrosis is the leading cause of organ dysfunction in diseases such as systemic sclerosis, liver cirrhosis, cardiac fibrosis, progressive kidney disease, and idiopathic pulmonary fibrosis. The hallmark of fibrosis is tissue remodeling with excess deposition of extracellular matrix components, predominantly collagens. Different cell types, cytokines, growth factors, and enzymes interact in complex pathogenic networks with myofibroblasts playing a pivotal role. MicroRNAs are small non-coding RNAs acting as negative regulators of gene expression at the post-transcriptional level. MicroRNAs have been associated with many basic cellular processes as well as with a wide spectrum of diseases, most notably cancer. This review provides a comprehensive overview of microRNAs regulating profibrotic pathways and extracellular matrix synthesis. The potential of miRNA for targeted therapeutic approaches in fibrotic disorders is also discussed.

Keywords: Fibrosis, fibroblasts, microRNA (miRNA)-mediated gene regulation regulation, transforming growth factor-beta (TGF-β), connective tissue growth factor (CTGF), extracellular matrix (ECM), epithelial-to-mesenchymal transition (EMT), signaling pathways, antagomirs..



INTRODUCTION

Fibrosis is a leading cause of organ dysfunction in diseases, either as the outcome of an uncontrolled reaction to chronic tissue injury, or as the primary disease itself in predisposed individuals [1-4] (Table 1). Independently from the initiating trigger and the tissue-specific response, the hallmark of fibrosis is excess deposition of extracellular matrix (ECM) components, predominantly collagens and associated glycoproteins. Accumulating ECM replaces functional tissue, and disrupts organ architecture [1]. Different cell types, cytokines, growth factors, and enzymes are orchestrated together in complex pathogenic networks, converging on the activation and/or recruitment of fibroblasts and their differentiation into myofibroblasts, which are α-smooth muscle actin (α-SMA) enriched cells overproducing ECM [5-7]. It is currently believed that along with these cellular key players, a number of regulatory factors also have substantial effects on fibrosis and may be responsible for inter-organ variability of fibrotic manifestations [8].

Table 1.

Examples of Primary (Idiopathic) and Secondary Fibrotic Disorders with Multiple/Single Organ Presentation


Primary Secondary
Systemic
Systemic sclerosis Graft versus host disease
Drug-induced nephrogenic systemic fibrosis
Sarcoidosis Secondary amyloidosis
Amyloidosis Post-radiation
Toxic environmental exposure
Storage disorders (hemochromatosis, glycogenosis, Gaucher’s disease, etc)
Lungs
Idiopathic pulmonary fibrosis Pneumoconiosis
Histiocytosis X Infectious pneumonitis
Cryptogenic organizing pneumonia Tuberculosis
Hypersensitive pneumonitis
Inherited disorders
Autoimmune diseases
Transplant rejection
Drug-induced
Post-radiation
Sarcoidosis
Amyloidosis
Heart
Idiopathic restrictive cardiomyopathy Coronary artery disease/myocardial infarction
Pressure-overload heart (long standing arterial hypertension, valvular disease)
Infectious myocarditis
Autoimmune diseases
Transplant rejection
Familial hypertrophic cardiomyopathy
Arrhythmogenic right ventricular cardiomyopathy
Drug-induced
Post-radiation
Sarcoidosis
Amyloidosis
Kidney
Idiopathic nephrotic syndrome Diabetic glomerulosclerosis
Idiopathic membranoproliferative glomerulonephritis Hypertensive nephrosclerosis
Autoimmune glomerular diseases
Drug-induced
Post-radiation
Amyloidosis
Transplant rejection
Liver
Primary biliary cirrhosis Chronic viral hepatitis
Primary sclerosing cholangitis Schistosomiasis
Alcoholic liver disease
Nonalcoholic fatty liver disease
Drug-induced
Toxic environmental exposure
Inherited metabolic disorders
Autoimmune hepatitis
Intestinal bypass
Table 2.

Major Molecular Mediators Elevated in Fibrotic Disorders


Cytokines
  Transforming growth factor-β (TGF-β)
  Interleukin (IL)-4
  IL-13
  IL-17
Chemokines
  CCL2
  CXCL8
  CXCL12
Growth Factors
  Connective tissue growth factor (CTGF)
  Platelet-derived growth factor (PDGF)
  Vascular endothelial growth factor (VEGF)
  Fibroblast growth factor (FGF)
  Insulin-like growth factor-1 (IGF-1)
Small Secreted Signaling Proteins
  Wnt proteins
  Endothelin-1 (ET-1)
Table 3.

MicroRNAs Linked to Fibrosis


MicroRNA Effect Organ Predicted Target and Confirmation Level Putative Role in Fibrosis References
let-7d Anti-fibrotic Lung Reporter gene assay (HMGA2) ↓EMT [48]
miR-107 Anti-fibrotic Kidney - ↓ Interstitial fibrosis in allograft transplant [49]
miR-132 Anti-fibrotic Heart, liver Target mRNA changes in mimic/silencing miRNA transfection (RAS-GTPase, MeCP2) ↑angiogenesis and ↓myofibroblasts differentiation [50, 51]
miR-133 Anti-fibrotic Heart Reporter gene assay (CTGF, TGF-β1) ↓ TGF-β signaling and ↓CTGF signaling [52, 53]
miR-141 Anti-fibrotic Kidney Reporter gene assay (TGF-β2) ↓TGF-β and ↓TGF-β dependent EMT [54]
miR-142-3p Pro-fibrotic Kidney - ↓Interstitial fibrosis in allograft transplant [49]
miR-15b, miR-16 Anti-fibrotic Liver, lung Reporter gene assay (Bcl-2) ↑apoptosis [55, 56]
miR-150 Anti-fibrotic Liver Target protein changes in mimic/silencing miRNA transfection (c-myb) ↓activation and proliferation in HSC [57]
miR-155 Pro-fibrotic Lung, liver Reporter gene assay (KGF-FGF-7); target protein changes in mimic/silencing miRNA transfection ↓mesenchymal-epithelial cross-talk [58, 59]
miR-17-92 cluster (miR-18a, 19a/b) Anti-fibrotic Liver, heart, lung Target protein changes in mimic/silencing miRNA transfection (CTGF; CTGF and TSP-1) ↓CTGF and ↓ TSP-1 [48, 60, 61]
miR-192 Pro-fibrotic Kidney Reporter gene assay (ZEB1/2) ↑collagen synthesis and ↑EMT [62, 63]
miR-194 (clustered with miR-215) Anti-fibrotic Liver Target protein changes in mimic/silencing miRNA transfection (rac1) ↓activation and ↓ proliferation in HSC [57]
miR-199a/b Pro-fibrotic Heart, Liver Reporter gene assay (Dyrk1); ↑ECM synthesis via calcineurin/NFAT signaling [64, 65]
miR-200 family (miR-200a/b) Anti-fibrotic Kidney, liver Reporter gene assay (TGFβ2 for miR-200a, ZEB1/2 for miR-200b); target mRNA changes in mimic/silencing miRNA transfection ↓TGF-β signaling and ↓ TGF-β -dependent EMT [54, 59, 66]
miR-204 Anti-fibrotic Kidney - ↓Interstitial fibrosis in allograft transplant [49]
miR-208 Pro-fibrotic Heart Reporter gene assay (THRAP1 and myostatin) ↓myocardial contractility and ↑fibrosis by de-repression of negative regulators of stress-response genes [67]
miR-21 Pro-fibrotic Lung, heart, kidney Reporter gene assay (Smad7 in lung); in vivo functional assay (Spry1 in heart); in vitro functional assay for Smad interactions ↑TGF-β canonical (via Smad7 de-repression of Smad2/3) and non canonical signaling (via Spry1 de-repression of ERK/MAPK) [68-70]
miR-211 Anti-fibrotic Kidney - ↓Interstitial fibrosis in allograft transplant [49]
miR-215 Pro-fibrotic Kidney Reporter gene assay (ZEB1/2) ↑EMT [63]
miR-216, miR-217 Pro-fibrotic Kidney Reporter gene assay (PTEN) ↑Akt signaling via PTEN de-repression [71]
miR-23a (clustered with miR-27a) Pro-fibrotic Lung Systems biology approach and real time PCR (ZEB1) ↑EMT [72]
miR-26a/b Anti-fibrotic Lung Microarray - [48]
miR-27b, miR-27a Pro-fibrotic Liver Reporter gene assay (RXR-α) ↑lipid accumulation and ↑proliferation of HSC [73]
miR-29a/b/c Anti-fibrotic SSc, heart, lung, kidney, liver Reporter gene assays (different collagens, FBN1, ELN1, MMP2, ITGB1; target mRNA changes in mimic/silencing miRNA transfection Directly ↓ECM synthesis [47, 48, 59, 74-77]
miR-30c Anti-fibrotic Heart, lung Reporter gene assay (CTGF) ↓CTGF signaling [48, 51]
miR-32 Pro-fibrotic Kidney - ↓Interstitial fibrosis in allograft transplant [49]
miR-335 Anti-fibrotic Liver Target mRNA changes in mimic/silencing miRNA transfection (TNC) ↓HSC activation and migration [78]
miR-338* Pro-fibrotic Lung Reporter gene assay (LPA1) ↑fibroblast proliferation [79]
miR-34a Pro-fibrotic Liver Reporter gene assay (ACSL1) ↑lipids biosynthesis [59, 80]
miR-377 (clustered with miR-382) Pro-fibrotic Kidney Reporter gene assay (PAK1, SOD) ↑fibronectin synthesis [81]
miR-382 Pro-fibrotic Kidney Reporter gene assay (SOD2) ↑EMT mediated by ↓ protection against mitochondrial oxidative stress [82]
miR-449a/b Anti-fibrotic Kidney Target mRNA changes in mimic/silencing miRNA transfection (SERPINE1) ↑ECM synthesis in hypoxic conditions via downregulation of miR-449 [83]
miR-590 Anti-fibrotic Heart Reporter gene assay (TGFβR2) ↓TGF-β signaling [53]

EMT = epithelial-to-mesenchymal transition; HMGA2 = high mobility group AT-hook 2; RAS-GTPase = Rat sarcoma guanosine triphosphatase; MeCP2 = methylCpG binding protein 2; CTGF = connective tissue growth factor; TGF-β = transforming growth factor- β; Bcl-2 = B-cell lymphoma 2; HSC = hepatic stellate cell; c-myb = avian myeloblastosis proto-oncoprotein; KGF = keratinocytes growth factor; ZEB = zinc finger E-box-binding homebox; rac1 = Ras-related C3 botulinum toxin substrate 1; Dyrk1 = dual-specificity tyrosine-(Y)-phosphorylation regulated kinase 1a; NFAT = nuclear factor of activated T-cells; βMHC = β myosin heavy chain; Smad = small mother against decapentaplegic; Spry1 = protein sprouty homolog 1; ERK = extracellular signal-regulated kinase; MAPK = mitogen activated protein kinase; PTEN = phosphatase and tensin homolog; Akt = non-specific serine/threonine-protein kinase; RXR-α = retinoid X receptor-α; FBN1 = fibrillin1; ELN1 = elastin1; MMP2 = metalloproteinase 2; ITGB1 = integrin β1; TNC = tenascin; LPA1 = lipophosphatidic acid; ACSL1 = Long-chain-fatty-acid-CoA ligase 1; PAK1 = p21 activated kinase 1; SOD = superoxide dismutase; TGF-β receptor 2 = transforming growth factor- β receptor type 2; TSP-1 = thrombospondin-1.

In this scenario, microRNAs (miRNAs) may play the role of conductors in the pathogenesis of fibrosis [9]. miRNAs are small, non-coding RNAs with a strictly regulated biogenesis. This is combined with an extremely flexible and sophisticated regulatory function, allowing simultaneous targeting of multiple proteins involved in different crucial biological pathways of specific cell types and tissues [10]. Consequently, miRNAs have been associated with many basic cellular processes such as cell death, proliferation and differentiation. On the other hand, miRNA dysregulation has also been linked to a wide spectrum of diseases, most notably cancer [11].

A number of recent reviews have addressed the role of miRNA in fibrosis with a focus on organ-specific miRNA alteration [12-15]. Here we paid special attention to miRNA alterations common to different fibrotic disorders such as systemic sclerosis (SSc) [16], liver cirrhosis (LC) [17], cardiac fibrosis (CF) [18, 19], chronic kidney disease (CKD) [20], and idiopathic pulmonary fibrosis (IPF) [21]. miRNAs directly regulating ECM synthesis and miRNAs regulating TGF-β/CTGF signaling as a key profibrotic pathway were considered for this review.

MicroRNA BIOGENESIS AND FUNCTION

miRNAs are a class of small, evolutionarily conserved, non-coding RNAs, which repress the expression of protein coding genes at the post-transcriptional level via a partially complementary binding to the 3’ untranslated region (UTR) of target mRNAs ([22], for comprehensive review). To date, 1426 miRNA encoding sequences have been identified in the human genome and more than half of them have been experimentally validated (ftp://mirbase.org/pub/mirbase/CU RRENT/genomes/hsa.gff) [23]. Most miRNA genes are located in introns, exons and UTRs of protein coding genes. Approximately one quarter of miRNAs are encoded in intergenic regions, and display an independent transcription unit, whereas intronic and exonic miRNAs are usually co-transcribed and co-expressed with the host gene [22]. In addition, a half of known miRNAs are encoded by polycistronic units, generating multiple miRNAs, often related in their structure and function [22, 24].

miRNAs are transcribed as long polyadenylated RNAs, pri-miRNAs, which contain one or more mature miRNA sequences. Pri-miRNAs are cut by the endonuclease Drosha and its co-factor DiGeorge syndrome critical region 8 (DGCR8), originating intermediate 60-100 nucleotide hairpin structures called pre-miRNAs [22]. Pre-miRNAs are transported to the cytoplasm by exportin 5 and further processed by another endonuclease, Dicer, releasing functionally active, mature miRNAs with a length of ≈ 22 nucleotides. Each mature miRNA is then incorporated into the RNA-induced silencing complex (RISC), which allows the recognition of target mRNA and the formation of a miRNA:mRNA duplex [22]. The base pairing of the duplex is mediated by a distinct seed region at nucleotides 2-8 of the miRNA, and alters mRNA stability or affects protein translation [25]. Translational repression and also target mRNA destabilization predominate in humans, although in rare cases miRNAs can switch their negative regulatory function towards gene activation by promoting mRNA translation [26].

The “imperfect” miRNA:mRNA binding is the basis for an extremely flexible regulatory system, as one miRNA can regulate hundreds of genes, and one gene containing multiple partially complementary binding sites in its 3’UTR can be regulated by multiple miRNAs [24]. In addition, this flexibility in miRNA:mRNA interaction is further expanded by a number of variables: miRNA binding site accessibility, RNA secondary structure, position of the binding site in the 3’UTR and its distance from the stop codon, and reciprocal interference between different miRNAs and RNA binding proteins sharing the same target [27]. As a consequence, miRNA activity depicts a gray scale rather than a black-or-white regulatory scenario [27, 28]. This aspect is particularly relevant for the regulation of multistep biological processes such as signaling pathways: while the effect of a given miRNA on the overall activity of the pathway can be potent, this often results from the sum of subtle modulations on multiple constituents of the same pathway rather than from strong effects on a single molecule [28]. Comprehensive in silico analysis of miRNA targets is provided at the miRGen database (http://www.diana.pcbi.upenn.edu/miRGen.html), which merges the results of multiple miRNA prediction databases, such as DIANA microT, miRanda (microrna.org), miRanda (miRBase), PicTar (4-way and 5-way), and TargetScanS [29].

Despite this versatile regulatory activity, the expression of miRNA genes is tightly controlled, and specific cell types often display a certain miRNA profile [22]. Regulation of miRNA expression at both the transcriptional and post-transcriptional level is still poorly understood. However, multiple mechanisms may play a role. At the transcriptional level, miRNAs can control their own expression establishing either positive or negative regulatory loops via an indirect regulation of transcriptional activators or repressors [30]. Epigenetic modifications such as DNA and histone methylation and histone deacetylation also participate in the modulation of miRNA transcription [31, 32]. At the post-transcriptional level, factors influencing the activity of the endonucleases Drosha and Dicer affect selective miRNA cluster maturation [33, 34]. In addition, some pseudogene mRNAs can act as miRNA decoy through a 3’UTR which is highly homologous to the one of the related protein-coding mRNA, thus modulating cell levels of “free” miRNA [35]. Finally, exosome-mediated cell-cell exchange of RNA has also been described as a determinant of cellular miRNA profile [36].

Fig. (1) gives an overview of miRNA biogenesis and function.

Fig. (1).

Schematic miRNA biogenesis and regulation. The miRNA gene is transcribed (pri-miRNA) and cut in the nucleus to generate pre-miRNA. Pre-miRNA is transported to the cytoplasm and is further processed there. Mature miRNA is originated as a mature miRNA:miRNA* (passenger strand) duplex. Mature miRNA is incorporated in RISC to bind a target mRNA. Perfect miRNA-mRNA binding leads to mRNA cleavage, while imperfect miRNA-mRNA binding leads to translational repression (rarely to translational stimulation). Levels of cellular bioactive miRNA can be controlled by both transcriptional regulation, including epigenetic modifications and target-dependent loops, and post-transcriptional regulation, including decoy mechanisms exerted by pseudogenes, and intercellular exosomedependent trafficking. DNMTs = DNA methyltransferases; HDACs = histone deacetylases; DGCR8 = DiGeorge syndrome critical region 8; RISC = RNA-induced silencing complex; ψ = pseudogene.


SSC: A MODEL FOR FIBROTIC DISEASES?

Despite fibrotic disorders include a large number of primary and secondary entities with distinct features, major extracellular mediators of fibrosis and related signaling pathways are nevertheless common to most of them. This includes cytokines, chemokines, growth factors, and other small secreted signaling proteins (Table 2) [2].

SSc is a systemic fibrotic disease of unknown origin characterized by the presence of three main pathophysiological pathways in each target organ/system: vascular injury, immune system activation, and interstitial fibrosis [37]. Thus, we propose SSc as a paradigm of fibrotic disorders, based on the following observations: i) fibrosis in SSc can virtually affect all organs/systems, although the disease displays apparent clinical involvement at preferential sites (skin, lungs, heart, kidneys, gastrointestinal tract, muscles); ii) initial factors triggering pro-fibrotic pathways in other fibrotic diseases are also discussed in SSc and SSc-like diseases including infections, autoimmune phenomena, and exposure to toxics or radiations [38-41]; iii) consistent evidence for the activation of major pro-fibrotic pathways common to most fibrotic diseases has been reported in SSc, including transforming growth factor-β (TGF-β) pathways, the platelet-derived growth factor (PDGF) pathway, and, most recently, the canonical Wnt-β-catenin pathway and the serotonin pathway [42-47].

OVERVIEW OF PRO- AND ANTI-FIBROTIC MicroRNAs

According to the current literature, ≈ 40 miRNAs have been linked to fibrosis in various organs and disease settings (Table 3). In general, the miRNA profile is specific for a given cell-type, and even more specific in relation to the developmental and functional organ status [22]. This might explain, why many miRNAs with pro- or anti-fibrotic properties were found to be dysregulated in specific organs and specific fibrotic diseases [48, 49, 52-54, 57, 62, 63, 67, 71-73, 78-83]. On the other hand, some miRNAs have been found to be abnormally expressed in more than one fibrotic disorder [47, 50, 51, 55, 56, 58-61, 64-66, 68-70, 74-77]. Predicted targets of approximately half of miRNAs involved in fibrosis have been validated by reporter gene assay [84]. Most of these miRNAs directly induce or protect from fibrosis by targeting TGF-β canonical and non-canonical pathways [53, 54, 68-71], connective tissue growth factor (CTGF) [51, 52, 60, 61], ECM structural proteins or enzymes involved in ECM remodeling [47, 74-77, 81, 83]. Another set of miRNAs indirectly regulates fibrogenesis by affecting epithelial-to-mesenchymal transition (EMT) [48, 50, 51, 58, 62, 63, 66, 82], or by inducing proliferation and resistance to apoptosis in myofibroblasts [51, 55-57, 73, 78, 80] (Table 3).

MicroRNA REGULATION OF TGF-B/CTGF SIGNALING

TGF-β is a central mediator of fibrosis [1] and it is ubiquitously expressed in tissues [85]. TGF-β also induces expression of CTGF, and CTGF can in turn enhance TGF-β signaling, along with a number of other pro-fibrotic factors including vascular endothelial growth factor (VEGF), endothelin-1 (ET), insulin-like growth factor-1 (IGF-1), integrins, and Wnts [86]. Therefore, miRNA regulation of TGF-β and CTGF signaling is of paramount interest. Up-to-date, 10 miRNAs showed consistent experimental evidence of being TGF-β/CTGF signaling regulators. Six of them were dysregulated in more than one fibrotic disorder (see Table 3).

miR-17-92 cluster. miR-17-92 is a proto-oncogenic cluster (also called oncomir-1) consisting of six miRNAs. Very recently, miR-18a and miR-19a/b from this cluster have been shown to regulate CTGF and thrombospondin-1 (TSP-1) in the context of liver and cardiac fibrosis [60, 61]. In a previous report, CTGF and TSP-1 were validated as direct miR-18a/miR-19a/b targets in cancer [87]. Kodama et al. [60] reported down-regulation of the miR-17-92 cluster in liver fibrosis in a p53-dependent manner and an associated up-regulation of CTGF. Similarly, van Almen et al. [61] showed down-regulation of the miR-17-92 cluster and spontaneous upregulation of CTGF and TSP-1 in a mouse model of age-related heart failure, which is characterized by widespread interstitial EMC accumulation. Down-regulation of the miR-17-92 cluster paralleled by CTGF and TSP-1 gene up-regulation was confirmed in human idiopathic cardiomyopathy specimens. Transfection experiments in aged neonatal rat cardiomyocytes were used to confirm CTGF and TSP-1 modulation by miR-18a and 19a/b. Down-regulation of the miR-17-92 cluster has also been reported in human IPF lung tissue [48].

miR-133, miR-30 and miR-590. Two other major regulators of CTGF expression in cardiac fibrosis are miR-133 and miR-30. miR-133 is a cardiac specific miRNA and its expression is limited to cardiomyocytes, while miR-30 is expressed in both cardiac fibroblasts and cardiomyocytes as well as in other tissues. Duisters et al. [52] reported miR-133- and miR-30c-mediated repression of CTGF in two animal models of overloaded heart disease: a rat renin-dependent hypertensive failing heart model (Ren2 rats) and a mouse mechanical stress-induced model (mice undergone transverse aortic constriction). They first showed down-regulation of the two miRNA families and up-regulation of CTGF in vivo, then confirmed the direct interaction of miR-133 and miR-30c with CTGF mRNA in vitro using cultured cardiomyocytes and cardiac fibroblasts, and finally proved the functional effects of this interaction on collagen synthesis. Interestingly, the authors also showed that miR-133 and miR-30c do not exert additive effects on CTGF regulation, which might be explained by the overlapping seed regions of these miRs in the 3’UTR of the CTGF gene. Similar down-regulation of miR-133 and miR-30c and increased levels of CTGF were observed in human hypertrophic heart.

The importance of miR-133 down-regulation in cardiac fibrosis is further supported by the findings of Shan and colleagues [53], who investigated the expression of miR-133 in the heart of smokers with chronic atrial fibrillation (AF), and in a canine model of nicotine-induced AF. Fibrotic structural remodeling was strongly induced by nicotine in both human and canine heart tissue. This was associated with an increase in TGF-β1 and the TGF-β pathway downstream targets CTGF and collagens both in vivo and in vitro. By in silico analysis, the authors selected miR-133 and miR-590 as potential regulators of TGF-β1 and TGF-βRII, respectively. miR-133 and miR-590 were strongly down-regulated in dog atrial fibroblasts treated with nicotine, and the direct interaction of the two miRNAs with their predicted targets could be confirmed by reporter gene assay. miR-30c down-regulation has also been reported in human IPF lung tissue [48].

miR-200. Most data link the five miR-200 family members to EMT [54, 66, 88], thus indicating an indirect role in fibrogenesis. Recent data from Wang et al. [54] showed in addition that the miR-200 family contributes directly to fibrosis by regulation of TGF-β2. Expression of miR-141 and miR-200a has been investigated in mouse models of early (ApoE knockout mice injected with streptozotocin) and advanced (C57BL6 mice treated with adenine) diabetic nephropathy, as well as in rat proximal tubular epithelial cells (NRK52E). Both miRNAs were down-regulated in mice, but only miR-200a was sensitive to TGF-β stimulation in NRK52E cells. At the same time, TGF-β stimulation induced transformation of NRK52E cells towards a mesenchymal phenotype as indicated by reduced E-cadherin expression and increased vimentin and α-SMA expression along with enhanced ECM synthesis. In turn, forced up-regulation of both miR-141 and miR-200a in NRK52E cells reduced TGF-β2, Smad3 activity and ECM proteins, and also prevented EMT, suggesting a miR-200a/TGF-β feedback loop in the pathogenesis of diabetic CKD. In addition, reporter gene assays confirmed TGF-β2 as a direct target of miR-141/miR-200a.

miR-21. Like the miR-17-92 cluster, miR-21 is traditionally regarded as an oncomir, and due to its ubiquitous expression it is one of the most widely investigated miRs [89]. Evidence for an important role of miR-21 in fibrosis comes from studies on heart, lung and kidney [68-70]. Thum and colleagues [69] described miR-21 up-regulation in three mouse models of heart failure: β1-adrenergic receptor transgenic mice, C57BL/6 mice undergone transverse aortic constriction, and C57BL/6 mice treated with isopropanol. In different cell types isolated from failing hearts, miR-21 was predominantly up-regulated in cardiac fibroblasts, and mediated protection from apoptosis. Anti-miR-21 treatment and extracellular signal-regulated kinase/mitogen activated protein kinase (ERK/MAPK) blockade induced apoptosis to a similar extent, suggesting a possible regulation of ERK/MAPK signaling by miR-21. Accordingly, overexpression of miR-21 in both mouse and rat fibroblasts led to increased expression and activation of ERK/MAPK proteins. The authors identified sprouty1, a negative regulator of ERK/MAPK, as the direct target of miR-21 mediating the effects on this pathway. Nevertheless, more recently, Patrick et al. [90] demonstrated in mir21-/- mice, which were exposed to different stimuli resulting in cardiac overload, that miR-21 contributes to, but is not essential for ERK/MAPK activation in stress-dependent cardiac remodeling.

Liu et al. [68] confirmed the pro-fibrotic activity of miR-21 in human IPF and murine bleomycin-induced lung fibrosis. Like in heart failure, miR-21 up-regulation was proportional to the severity of lung fibrosis in the animal model. TGF-β1 expression showed the same trend. Therefore, the authors elucidated the causal relation between miR-21 and TGF-β1, showing that miR-21 expression was reduced in TGFβRII dominant negative mice injected with bleomycin. Consistently, TGF-β1 treatment of cultured pulmonary fibroblasts induced miR-21 up-regulation and, in turn, miR-21 overexpression induced TGF-β1 expression, Smad2 activation, and ECM synthesis. These effects were mediated by direct repression of Smad7, the inhibitory Smad which counter-regulates Smad2/3 activation. Finally, administration of miR-21 antisense oligonucleotides in vivo attenuated bleomycin-induced lung fibrosis.

Studies on the role of miR-21 in renal fibrosis further elucidated the interaction of this miRNA with TGF-β/Smad signaling [70]. In these experiments, TGF-β-mediated upregulation of miR-21 in tubular epithelial cells was prevented by knockdown of Smad3, but not by knockdown of Smad2. Consistently, Smad3 deficient mice, but not Smad2 deficient mice, exposed to kidney injury by unilateral urethral obstruction were protected from miR-21 up-regulation and fibrosis.

miR-216, miR-217 and miR-192. miR-216 and miR-217 are two clustered miRNAs encoded in the second intron of the RP23 gene. They showed pro-fibrotic effects in murine mesangial cells (MCs) [71]. Kato et al. reported upregulation of miR-216/217 in mouse MCs following TGF-β treatment, with consequent amplification of TGF-β signaling. miR-216 and miR-217 directly target phosphatase and tensin homolog (PTEN), which inhibits the phosphoinositide-3-kinase pathway (PI3K), thereby preventing activation of Akt. Thus, down-regulation of PTEN de-represses Akt signaling in MCs leading to prolonged cell survival, hypertrophy and increased collagen synthesis. In addition, since the promoter region of the RP23 gene contains multiple Smad binding elements and E-boxes, the authors could demonstrate that the expression of miR-216 and miR-217 in MCs was induced by TGF-β and zinc finger E-box-binding homebox (Zeb) 1 and 2. In turn, Zeb 1 and 2 are regulated by miR-192 [62].

MicroRNA REGULATION OF ECM PROTEINS

miR-29. miR-29 is the best characterized direct regulator of ECM synthesis. Its involvement in fibrosis as an anti-fibrotic miRNA has been reported in SSc as well as in other major fibrotic disorders. The miR-29 family includes three members: miR-29a/b/c. We could show that all three miRNAs are down-regulated in human SSc dermal fibroblasts and SSc skin [47]. miR-29a showed the strongest down-regulation in tissues and cells, and the most consistent functional effects on the expression of collagen type I and type III. In addition, COL3A1 was validated as a direct target of miR-29a. We also demonstrated that PDGF-B and TGF-β down-regulate miR-29a, and that down-regulation of miR-29a leads to further upregulation of PDGF-B and TGF-β. This creates a positive feed-back loop among profibrotic mechanisms leading to the uncontrolled release of extracellular matrix proteins in SSc. Results were confirmed in a bleomycin-induced mouse model of skin fibrosis, in which the tyrosine-kinase inhibitor imatinib mesylate was able to restore miR-29a levels.

In murine and human hearts, down-regulation of the miR-29 family following myocardial infarction has been reported [74]. Van Rooji et al. showed that cardiac fibroblasts, but not cardiomyocytes, are responsible for the effects of miR-29 on ECM synthesis in myocardial infarction. They hypothesized that miR-29 down-regulation is mediated by TGF-β following cardiac injury, and that this axis is controlled by the parallel induction of brain natriuretic peptide (BNP), which causes repression of TGF-β. COL1A1, COL1A2, COL3A1, FBN1, and ELN1 genes were validated as direct miR-29 targets. In addition, miR-29b knockdown with cholesterol-conjugated antagomirs in C57BL/6 male mice induced COL1A1, COL1A2, and COL3A1 mRNA overexpression in cardiac tissue.

A clear anti-fibrotic function of the miR-29 family was also shown in hypertensive kidney disease [76]. Liu et al. found that miR-29b was overexpressed in a mouse model, which is protected against salt-induced hypertensive renal medullary fibrosis (SS-13BN) after high-salt diet. Silencing of miR-29b in the kidney of these animals caused up-regulation of a large number of ECM and ECM-modulating genes, subsequently validated as direct targets of miR-29b by reporter gene assay (COL1A1, COL3A1, COL4A1, COL5A1, COL5A2, COL5A3, COL7A1, COL8A1, MMP2, and ITGB1). As reported by Du et al. [77], miR-29a down-regulation can also be induced in vitro in human proximal tubular epithelial cells (HK-2 cells) by high glucose and TGF-β stimulation, with similar effects on the expression of multiple collagen genes, suggesting a role for miR-29 also in diabetic nephropathy.

Regarding lung fibrosis, down-regulation of miR-29 has been reported by Cushing et al. [75] in bleomycin-treated mice, which was paralleled by an increase in COL3A1 and COL4A1 expression. In addition, the same authors investigated the effects of silencing of miR-29 family members in human fetal lung fibroblasts. They identified new direct targets, among them both structural ECM genes and enzymes involved in tissue remodeling, namely NID1, ITGA11, ADAMTS9, and ADAMTS12. In lungs as well as in the skin, heart and kidney, TGF-β was able to reduce miR-29 levels.

Lastly, miR-29 down-regulation (miR-29c) has been reported in a mouse model of non-alcoholic steatohepatitis [59], although functional investigations of miR-29 in the liver are still lacking.

Other miRNAs. Additional miRNAs have been shown to regulate ECM synthesis in the kidney: miR-377 and miR-449. Wang et al. [81] showed pro-fibrotic effects of miR-377 in human mesangial cells following high glucose and TGF-β administration and in animal models of type I diabetes. These effects were mediated by an indirect upregulation of fibronectin via targeting the fibronectin inhibitors p21-activated kinase and superoxide dismutase. Muth et al. [83] demonstrated that the expression of miR-449a/b was reduced in human fibroblasts cultured under hypoxic conditions and in human kidney transplants displaying chronic allograft nephropathy. miR-449 down-regulation induced Serpine-1 (PAI-1) expression. Serpine-1 inhibits tissue-type (tPA) and urokinase-type plasminogen activator (uPA), which mediate proteolysis of ECM proteins and activation of latent metalloproteinases (MMPs). Thus, the authors hypothesized that miR-449 down-regulation promotes ECM accumulation in hypoxic fibroblasts and chronic post-transplant allograft remodeling in the kidney.

CONCLUSIONS AND PERSPECTIVES

Our review outlines the potentially important role of miRNAs in fibrosis. With some exceptions, most of the miRNAs reported in this review are ubiquitously expressed, indicating that the majority of pro- or anti-fibrotic miRNAs might be common regulators of fibrosis in different organs and disease settings. In addition, several key pro-fibrotic molecules are regulated by multiple miRNAs [51-54, 60, 61, 68-71].

The partially redundant interaction between miRNAs and their targets might hamper the use of miRNA mimics or inhibitors as potential therapeutic tools. However, data on the use of antagomirs in animal models show promising results [68, 74, 90]. It is important for the development of targeted therapies that miRNAs exert rather mild to moderate effects on single molecules, but that these effects are additive when multiple molecules of a certain pathophysiological pathway are regulated. Therefore, miRNAs are of particular interest for targeted therapies in fibrosis, when they regulate multiple rate limiting molecules of profibrotic pathways. In addition, since differentially expressed miRNAs are often disease specific and do not play major roles under normal conditions in adult tissue or in quiescent cells, targeting of an aberrantly expressed miRNA is expected to be well tolerated [91]. Indeed, many miRNA knockout animal models do not display a specific phenotype unless they are challenged with a stimulus causing cellular stress in target organs [69]. However, the miRNA machinery in fibrosis is still far from being fully elucidated, although research in this field in the last years has been considerably fostered.

ACKNOWLEDGEMENT

Supported by a grant from the EULAR ODP program.

CONFLICT OF INTEREST

O. Distler had consultancy relationship and/or has received research funding from Actelion, Pfizer, Ergonex, BMS, Sanofi-Aventis, United BioSource Corporation, medac, Biovitrium, Boehringer Ingelheim Pharma, Novartis, 4 D Science, Bayer and Active Biotec in the area of potential treatments of scleroderma and its complications, he is holding a patent on miR-29 for the treatment of scleroderma.

REFERENCES

[1] Wynn TA. Cellular and molecular mechanisms of fibrosis J Pathol 2007; 214: 199-210.
[2] Wynn TA. Common and unique mechanisms regulate fibrosis in various fibroproliferative diseases J Clin Invest 2007; 117: 524-9.
[3] Zimmer V, Lammert F. Genetics in liver disease: new concepts Curr Opin Gastroenterol 2011; 27: 231-9.
[4] Romano E, Manetti M, Guiducci S, Ceccarelli C, Allanore Y, Matucci CM. The genetics of systemic sclerosis: an update Clin Exp Rheumatol 2011; 29: S75-86.
[5] Tomasek JJ, Gabbiani G, Hinz B, Chaponnier C, Brown RA. Myofibroblasts and mechano-regulation of connective tissue remodeling Nat Rev Mol Cell Biol 2002; 3: 349-63.
[6] Quan TE, Cowper SE, Bucala R. The role of circulating fibrocytes in fibrosis Curr Rheumatol Rep 2006; 8: 145-50.
[7] Kalluri R, Neilson EG. Epithelial-mesenchymal transition and its implications for fibrosis J Clin Invest 2003; 112: 1776-84.
[8] Mehal WZ, Iredale J, Friedman SL. Scraping fibrosis Nat Med 2011; 17: 552-3.
[9] Chau BN, Brenner DA. What goes up must come down: the emerging role of microRNA in fibrosis Hepatology 2011; 53: 4-6.
[10] Liu N, Olson EN. MicroRNA regulatory networks in cardiovascular development Dev Cell 2010; 18: 510-25.
[11] Kloosterman WP, Plasterk RH. The diverse functions of microRNAs in animal development and disease Dev Cell 2006; 11: 441-50.
[12] van Rooji E, Olson EN. Searching for MIR-acles in cardiac fibrosis Circ Res 2009; 104: 138-40.
[13] Jiang X, Tsitsiou E, Herrick SE, Lindsay M. MicroRNAs and the regulation of fibrosis FEBS J 2010; 277: 2015-1.
[14] Pandit KV, Milosevic J, Kaminski N. MicroRNAs in idiopathic pulmonary fibrosis Transl Res 2011; 157: 191-9.
[15] Lorenzen JM, Haller H, Thum T. MicroRNAs as mediators and therapeutic targets in chronic kidney disease Nat Rev Nephrol 2011; 7: 286-94.
[16] Wei J, Bhattacharyya S, Tourtellotte WG, Varga J. Fibrosis in systemic sclerosis: emerging concepts and implications for targeted therapy Autoimmun Rev 2011; 10: 267-75.
[17] Pinzani M, Rosselli M, Zuckermann M. Liver cirrhosis Best Pract Res Clin Gastroenterol 2011; 25: 281-90.
[18] Krenning G, Zeisberg EM, Kalluri R. The origin of fibroblasts and mechanisms of cardiac fibrosis J Cell Physiol 2010; 225: 631-7.
[19] Creemers EE, Pinto YM. Molecular mechanisms that control interstitial fibrosis in the pressure-overloaded heart Cardiovasc Res 2011; 89: 265-82.
[20] Dussaule JC, Guerrot D, Huby AC, et al. The role of cell plasticity in progression and reversal of renal fibrosis Int J Exp Pathol 2011; 92: 151-7.
[21] Wynn TA. Integrating mechanisms of pulmonary fibrosis J Exp Med 2011; 208: 1339-50.
[22] Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function Cell 2004; 116: 281-97.
[23] Kozomara A, Griffiths JS. miRBase: integrating microRNA annotation and deep-sequencing data Nucleic Acids Res 2011; 39: D152-7.
[24] Ambros V. The functions of animal microRNAs Nature 2004; 431: 350-5.
[25] Filipowicz W, Bhattacharyya SN, Sonenberg N. Mechanisms of post-transcriptional regulation by microRNAs: are the answers in sight? Nat Rev Genet 2008; 9: 102-4.
[26] Vasudevan S, Tong Y, Steitz JA. Switching from repression to activation: microRNAs can up-regulate translation Science 2007; 318: 1931-4.
[27] Bartel DP. MicroRNAs: target recognition and regulatory functions Cell 2009; 136: 215-33.
[28] Inui M, Martello G, Piccolo S. MicroRNA control of signal transduction Nat Rev Mol Cell Biol 2010; 11: 252-63.
[29] Megraw M, Sethupathy P, Corda B, Hatzigeorgiou AG. miRGen: a database for the study of animal microRNA genomic organization and function Nucleic Acids Res 2007; 35: D149-55.
[30] Chen CY, Chen ST, Fuh CS, Juan HF, Huang HC. Coregulation of transcription factors and microRNAs in human transcriptional regulatory network BMC Bioinformatics 2011; 12: S41.
[31] Vrba L, Garbe JC, Stampfer MR, Futscher BW. Epigenetic regulation of normal human mammary cell type specific miRNAs Genome Res 2011; 21: 2026-37.
[32] Lee S, Jung JW, Park SB, et al. Histone deacetylase regulates high mobility group A2-targeting microRNAs in human cord blood-derived multipotent stem cell aging Cell Mol Life Sci 2011; 68: 325-36.
[33] Davis BN, Hilyard AC, Nguyen PH, Lagna G, Hata A. Smad protein bind a conserved RNA sequence to promote microRNA maturation by Drosha Mol Cell 2010; 39: 373-84.
[34] Boominathan L. The tumor suppressors p53, p63, and p73 are regulators of microRNA processing complex PLoS One 2010; 5: e10615.
[35] Poliseno L, Salmena L, Zhang J, Carver B, Haveman WJ, Pandolfi PP. A coding-independent function of gene and pseudogene mRNAs regulates tumor biology Nature 2010; 465: 1033-40.
[36] Valadi H, Ekström K, Bossios A, Sjöstrand M, Lee JJ, Lötval JO. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells Nat Cell Biol 2007; 9: 654-9.
[37] Varga J, Denton CP. Scleroderma, inflammatory myopathies, and overlap syndromes In: Firestein GS, Budd RC, Harris ED, Jr, McInnes IB, Ruddy S, Sergent JS, Eds. Kelley’s Textbook of Rheumatology. 8th. USA: Saunders Elsevier 2009; pp. 1311-51.
[38] Markiewicz M, Smith EA, Rubinchik S, Dong JY, Trojanowska M, LeRoy EC. The 72-kilodalton IE-1 protein of human cytomegalovirus (HCMV) is a potent inducer of connective tissue growth factor (CTGF) in human dermal fibroblasts Clin Exp Rheumatol 2004; 22: S31-4.
[39] Hasegawa M, Fujimoto M, Takehara K, Sato S. Pathogenesis of systemic sclerosis: altered B cell function is the key linking systemic autoimmunity and tissue fibrosis J Dermatol Sci 2005; 39: 1-7.
[40] Garabrant DH, Lacey JV Jr, Laing TJ, et al. Scleroderma and solvent exposure among women Am J Epidemiol 2003; 157: 493-500.
[41] Varga J, Haustein UF, Creech RH, Dwyer JP, Jimenez SA. Exaggerated radiation-induced fibrosis in patients with systemic sclerosis JAMA 1991; 265: 3292-5.
[42] Ishida W, Mori Y, Lakos J, et al. Intracellular TGF-β blockade abrogates Smad-dependent fibroblast activation in vitro and in vivo J Invest Dermatol 2006; 126: 1733-44.
[43] Pannu J, Gore HE, Yamanaka M, et al. An increased transforming growth factor β receptor type I: type II ratio contributes to elevated collagen protein synthesis that is resistant to inhibition via a kinase-deficient transforming growth factor β receptor type II in scleroderma Arthritis Rheum 2004; 50: 1566-77.
[44] Svegliati S, Cancello R, Sambo P, et al. Platelet-derived growth factor and reactive oxygen species (ROS) regulate RAS protein levels in primary human fibroblasts via ERK1/2. Amplification of ROS and RAS in systemic sclerosis fibroblasts J Biol Chem 2005; 280: 36474-82.
[45] Bergmann C, Akhmetshina A, Dees C, et al. Inhibition of glycogen synthase kinase3{beta} induces dermal fibrosis by activation of the canonical Wnt pathway Ann Rheum Dis 2011; 70: 2191-8.
[46] Dees C, Akhmetshina A, Zerr P, et al. Platelet-derived serotonin links vascular disease and tissue fibrosis J Exp Med 2011; 208: 961-72.
[47] Maurer B, Stanczyck J, Jungel A, et al. MicroRNA-29, a key regulator of collagen expression in systemic sclerosis Arthritis Rheum 2010; 62: 1733-43.
[48] Pandit KV, Corcoran D, Yousef H, et al. Inhibition and role of let-7d in idiopathic pulmonary fibrosis Am J Respir Crit Care Med 2010; 182: 220-9.
[49] Scian MJ, Maluf DG, David KG, et al. MicroRNA profiles in allograft tissues and paired urine associate with chronic allograft dysfunction with IF/TA Am J Transplant 2011; 11: 2110-2.
[50] Katare R, Riu F, Mitchell K, et al. Transplantation of human pericyte progenitor cells improves the repair of infarcted heart through activation of an angiogenic program involving micro-RNA-132 Circ Res 2011; 109: 894-906.
[51] Mann J, Chu DC, Maxwell A, et al. MeCP2 controls an epigenetic pathway that promotes myofibroblasts transdifferentiation and fibrosis Gastroenterology 2010; 138: 705-14.
[52] Duisters RF, Tijsen AJ, Schroen B, et al. miR-133 and miR-30 regulate connective tissue growth factor: implications for a role of microRNAs in myocardial matrix remodeling Circ Res 2009; 104: 170-8.
[53] Shan H, Zhang Y, Lu Y, et al. Down-regulation of miR-133 and miR-590 contributes to atrial remodelling in canines Cardiovasc Res 2009; 83: 465-72.
[54] Wang B, Koh P, Winbanks C, et al. miR-200a prevents renal fibrogenesis through repression of TGF-β2 expression Diabetes 2011; 60: 280-7.
[55] Guo CJ, Pan Q, Li DG, Sun H, Liu BW. miR-15b and miR-16 are implicated in activation of the rat hepatic stellate cell: an essential role for apoptosis J Hepatol 2009; 50: 766-8.
[56] Ezzie ME, Crawford M, Choo JH, et al. Gene expression network in COPD: microRNA and mRNA regulation Thorax 2012; 67: 122-31.
[57] Venugopal SK, Jiang J, Kim TH, et al. Liver fibrosis causes down-regulation of miR-150 and miR-194 in hepatic stellate cells, and their overexpression causes decreased stellate cell activation Am J Physiol Gastrointest Liver Physiol 2010; 298: G101-6.
[58] Pottier N, Maurin T, Chevalier B, et al. Indentification of keratinocyte growth factor as a target of miR-155 in lung fibroblasts: implication in epithelial-mesenchymal interactions PLoS One 2009; 4: e6718.
[59] Pogribny IP, Starlard DA, Tryndyak VP, et al. Difference in expression of hepatic miR-29c, miR-34a, miR-155, and miR-200b is associated with strain-specific susceptibility to dietary nonalcoholic steatohepatitis in mice Lab Invest 2010; 90: 1437-46.
[60] Kodama T, Takehara T, Hikita H, et al. Increases in p53 expression induce CTGF synthesis by mouse and human hepatocytes and result in liver fibrosis in mice J Clin Invest 2011; 121: 3343-56.
[61] van Almen GC, Verhesen W, van Leeuwen RE, et al. MicroRNA- 18 and microRNA-19 regulate CTGF and TSP-1 expression in age-related heart failure Aging Cell 2011; 10: 769-9.
[62] Kato M, Zhang J, Wang M, et al. MicroRNA-192 in diabetic kidney glomeruli and its function in TGF-beta-induced collagen expression via inhibition of E-box repressors Proc Natl Acad Sci USA 2007; 104: 3432-7.
[63] Wang B, Herman EM, Koh P. E-cadherin expression is regulated by miR-192/215 by a mechanism that is independent of the profibrotic effects of transforming growth factor-beta Diabetes 2010; 59: 1794-802.
[64] da Costa Martins PA, Salic K, et al. MicroRNA-199b targets the nuclear kinase Dyrk1a in an auto amplification-loop promoting calcineurin-NFAT signaling Nat Cell Biol 2010; 12: 1220-7.
[65] Murakami Y, Toyoda H, Tanaka M, et al. The progression of liver fibrosis is related to overexpression of miR-199 and miR-200 families PLoS One 2011; 6: e16081.
[66] Oba S, Kumano S, Suzuki E, et al. miR-200b precursor can ameliorate renal tubulointerstitial fibrosis PLoS One 2010; 5: e13614.
[67] van Rooji E, Sutherland LB, Qi X, Richardson JA, Hill J, Olson EN. Control of stress-dependent cardiac growth and gene expression by a microRNA Science 2007; 316: 575-9.
[68] Liu G, Friggeri A, Yang Y, et al. miR-21 mediates fibrogenic activation of pulmonary fibroblasts and lung fibrosis J Exp Med 2010; 207: 1589-97.
[69] Thum T, Gross C, Fiedler J, et al. MIcroRNA-21 contributes to myocardial disease by stimulating MAP kinase signalling in fibroblasts Nature 2008; 456: 980-4.
[70] Zhong X, Chung AC, Chen XY, Meng XM, Lan HY. Smad3-mediated up-regulation of miR-21 promotes renal fibrosis J Am Soc Nephrol 2011; 22: 1668-81.
[71] Kato M, Putta S, Wang M, et al. TGF-beta activates Akt kinase through a microRNA-dependent amplifying circuit targeting PTEN Nat Cell Biol 2009; 11: 881-9.
[72] Cho JH, Gelinas R, Wang K, et al. Systems biology of interstitial lung disease: mRNA and microRNA expression changes BMC Med Genomics 2011; 4: 8.
[73] Ji J, Zhang J, Huang G, Qian J, Wang X, Mei S. Over-expressed miRNA-27a and 27b influence fat accumulation and cell proliferation during rat hepatic stellate cell activation FEBS Lett 2009; 583: 759-66.
[74] van Rooji E, Sutherland LB, Thatcher JE, et al. Dysregulation of microRNAs after myocardial infarction reveals a role of mIR-29 in cardiac fibrosis Proc Natl Acad Sci USA 2008; 105: 13027-32.
[75] Cushing L, Kuang PP, Qian J, et al. miR-29 is a major regulator of genes associated with pulmonary fibrosis Am J Respir Cell Mol Biol 2011; 45: 287-94.
[76] Liu Y, Taylor NE, Lu L, et al. Renal medullary microRNAs in Dahl salt-sensitive rats: miR-29b regulates several collagens and related genes Hypertension 2010; 55: 974-82.
[77] Du B. High glucose down-regulates miR-29a to increase collagen IV production in HK-2 cells FEBS Lett 2010; 584: 811-6.
[78] Chen C, Wu CQ, Zhang ZQ, Yao DK, Zhu L. Loss of expression of miR-335 is implicated in hepatic stellate cell migration and activation Exp Cell Res 2011; 317: 1714-25.
[79] Zhang H, Liu X, Chen S, et al. Tectorigenin inhibits the in vitro proliferation and enhances miR-338* expression of pulmonary fibroblasts in rats with idiopathic pulmonary fibrosis J Ethnopharmacol 2010; 131: 165-73.
[80] Li WQ, Chen C, Xu MD, et al. The rno-miR-34 family is upregulated and targets ACSL1 in dimethylnitrosamine-induced hepatic fibrosis in rats FEBS J 2011; 278: 1522-32.
[81] Wang Q, Wang Y, Minto AW, Wang J, Shi Q, Li X. MicroRNA-377 is up-regulated and can lead to increased fibronectin production in diabetic nephropathy FASEB J 2008; 22: 4126-35.
[82] Kriegel AJ, Fang Y, Liu Y, et al. MicroRNA-target pairs in human renal epithelial cells treated with transforming growth factor β1: a novel role of miR-382 Nucleic Acids Res 2010; 38: 8338-47.
[83] Muth M, Theophile K, Hussein K, Jacobi C, Kreipe H, Bock O. Hypoxia-induced down-regulation of microRNA-449a/b impairs control over targeted SERPINE-1 (PAI-1) mRNA – a mechanism involved in SERPINE-1 (PAI-1) overexpression J Transl Med 2010; 8: 33.
[84] Nicolas FE. Experimental validation of microRNA targets using a luciferase reporter system Methods Mol Biol 2011; 732: 139-52.
[85] Santibanez JF, Quintanilla M, Bernabeu C. TGF-β/ TGF-β receptor system and its role in physiological and pathological conditions Clin Sci 2011; 121: 233-51.
[86] Leask A, Abraham DJ. All in the CCN family: essential matricellular signaling modulators emerge from the bunker J Cell Sci 2006; 119: 4803-10.
[87] Dews M, Homayouni A, Yu D, et al. Augmentation of tumor angiogenesis by a Myc-activated microRNA cluster Nat Gen 2006; 38: 1060-5.
[88] Gregory PA, Bert AG, Paterson EL, et al. The miR-200 family and miR-205 regulate epithelial to mesenchymal transition by targeting ZEB1 and SIP1 Nat Cell Biol 2008; 10: 593-601.
[89] Kumarswamy R, Volkmann I, Thum T. Regulation and function of miR-21 in health and disease RNA Biol 2011; 8: 1-8.
[90] Patrick DM, Montgomery RL, Qi X, Obad S, Kauppinen S, Hill JA. Stress-dependent cardiac remodeling occurs in the absence of microRNA-21 in mice J Clin Invest 2010; 120: 3912-6.
[91] Broderick JA, Zamore D. MicroRNA therapeutics Gene Ther 2011; 18: 1104-0.