Abstract
Purpose of Review
Myofibroblasts are the fundamental drivers of fibrosing disorders; there is great value in better defining epigenetic networks involved in myofibroblast behavior. Complex epigenetic paradigms, which are likely organ and/or disease specific, direct pathologic myofibroblast phenotypes. In this review, we highlight epigenetic regulators and the mechanisms through which they shape myofibroblast phenotype in fibrotic diseases of different organs.
Recent Findings
Hundreds of genes and their expression contribute to the myofibroblast transcriptional regime influencing myofibroblast phenotype. An increasingly large number of epigenetic modifications have been identified in the regulation of these signaling pathways driving myofibroblast activation and disease progression. Drugs that inhibit or reverse profibrotic epigenetic modifications have shown promise in vitro and in vivo; however, no current epigenetic therapies have been approved to treat fibrosis. Newly described epigenetic mechanisms will be mentioned, along with potential therapeutic targets and innovative strategies to further understand myofibroblast-directed fibrosis.
Summary
Epigenetic regulators that direct myofibroblast behavior and differentiation into pathologic myofibroblast phenotypes in fibrotic disorders comprise both overlapping and organ-specific epigenetic mechanisms.
Keywords: Myofibroblast, epigenetic, fibroblast, fibrosis
I. Introduction
Myofibroblasts are considered the primary cell type responsible for normal wound healing and tissue injury repair. Excessive or aberrant activation of repair processes, however, often leads to undesired, pathologic fibrosis (scarring) with subsequent organ dysfunction, and can cause premature mortality. In addition to fibrosis of specific tissues, myofibroblasts also feature prominently in diffuse disease processes such as systemic sclerosis (scleroderma). Furthermore, fibrosis provides an environment in which cancer is more likely to develop, as in hepatocellular carcinoma [1]. Myofibroblasts themselves have been shown to play a role in tumor development and invasion [2]. Despite the prominent role of fibrosis in diseases affecting many different organs, only two specific antifibrotic drugs are FDA-approved for idiopathic pulmonary fibrosis. Most clinicians have used anti-inflammatory or immunomodulatory agents to treat fibrosis, with mixed results. Therefore, elucidating the molecular etiologies for the activation of myofibroblasts is of great interest to researchers and clinicians. Epigenetic mechanisms are the principal regulators of cell phenotype or “identity,” and their role in the differentiation of myofibroblasts has been studied intensely but remains far from complete. As the discovery and development of epigenetic-directed therapies advance and the potential of epigenetic editing becomes reality, an improved understanding of the epigenetic regulation of myofibroblasts will provide promising new therapeutic targets to prevent, stop, or even reverse, fibrosis.
II. Myofibroblast-Origin, fibrotic phenotype, and drivers
Origin
The origin of myofibroblasts seems to be quite diverse, and can be organ or disease specific. Fibroblasts, smooth muscle cells, pericytes, mesenchymal stem/progenitor cells, bone marrow derived cells, adipose tissue cells, and epithelial and endothelial cells via epithelial-to-mesenchymal transition (EMT) and endothelial-to-mesenchymal transition (endoMT) have all been described as precursors, which can transdifferentiate into myofibroblasts when appropriately stimulated. Myofibroblasts have both fibroblast secretory and smooth muscle contractile characteristics. The myofibroblast phenotype is typified by the expression of smooth muscle α-actin (SMA) and the secretion of multiple pro-fibrogenic mediators such as TGFβ1, type I/III collagen, and tissue inhibitor of metalloproteinase-1 (TIMP-1), a collagenase inhibitor. SMA strengthens activated myofibroblast contractile activity and is commonly used as a myofibroblast marker though there is no known single marker to discriminate myofibroblasts, just as there are no specific markers to distinguish fibroblasts in general from other mesenchymal cells [3].
Myofibroblast Phenotype in Fibrosis
Activated myofibroblasts are the fundamental drivers of fibrosing disorders and there is great clinical value in better defining myofibroblast origin, fate, and characteristic markers and proteins. In response to injury or infection, the appearance and proliferation of myofibroblast results in deposition of collagen-rich extracellular matrix (ECM). Myofibroblast responses are attenuated as the repair process forms a scar in normal wound healing. They are cleared predominantly through apoptosis or the cells revert back to a more dormant fibroblast phenotype. Pathologic fibrosis results if active myofibroblasts persist in tissues, with uncontrolled proliferation and excessive collagen deposition. It has been increasingly demonstrated that epigenetic mechanisms contribute prominently to this “hyperactive” phenotype [3].
Myofibroblasts are also the principal cell type involved in tumor stroma remodeling as they establish a microenvironment for epithelial tumor cells to thrive and drive cancer progression [2]. The epigenetic regulation of cancer-related myofibroblasts is not included in this review, but is an immensely important topic that should be considered separately.
Myofibroblast Drivers
Transforming growth factor β1 (TGFβ1) drives profibrotic myofibroblast differentiation from precursor cells and stimulates expression of extracellular matrix and other genes essential to the pathogenesis of fibrosis. TGFβ1 induces fibrosis by activation of canonical (Smad-based) and non-canonical (non-Smad-based) signaling pathways. Downstream, complex interactions regulate myofibroblast activation and fibrogenesis. Multiple cytokines can suppress TGFβ1-mediated fibrogenesis such as basic fibroblast growth factors (bFGF, FGF-2), interferon gamma (IFNγ), and Interleukin-1 (IL-1). Anti-fibrogenic antagonists of TGFβ1 generally downregulate myofibroblast SMA expression but others have also been shown to decrease collagen deposition and contraction and induce apoptosis [3].
Another pivotal myofibroblast activator is mechanical stress or stiffness of extracellular matrix. ECM strain and stiffness, generated in part by myofibroblast contraction, can be sensed by other mechanosensitive cells, and positively feed back to sustain profibrotic activation. This topic was well reviewed by Hinz in 2016 [3]. Along with TGFβ1 and mechanical stress, a myriad of other factors can also activate myofibroblasts. A list of recent publications on myofibroblast-inducing factors and an overall review on current myofibroblast literature is available in the aforementioned review [3]. A dominant transcription factor or group of regulatory factors has not be identified to globally direct MTD. Hundreds of genes and their expression can contribute to the myofibroblast transcriptional regime and influence MTD. How these are coordinately regulated to program myofibroblast appearance and behavior is not completely understood, but epigenetic mechanisms are thought to play a prominent role.
III. Epigenetic Regulation of Myofibroblasts
Epigenetic modifications, heritable changes in the genome that do not alter the DNA sequence, influence and regulate gene expression. Epigenetic mechanisms include stable chromatin modifications and interactions, changes in DNA or protein conformation, nuclear organization, and non-coding RNAs. These mechanisms interact in multiple ways to allow or restrict access of transcription factors, transcriptional machinery, and epigenetic modifiers to the DNA sequence, thus affecting promoter and enhancer function, and gene transcription. Non-coding RNAs have predominantly post-transcriptional effects, but also modify the function of other epigenetic modifiers.
DNA Methylation
The most common epigenetic change associated with gene silencing is methylation of 5-cytosine (5meC) at CpG islands at promoter sites by DNA methyltransferases (DNMTs). Inhibition of gene transcription may occur by direct blockage of transcription factor binding to methylated CpG sites, or indirectly through proteins such as methyl CpG binding protein (MeCP2). MeCP2 binds to methylated DNA through a methyl CpG-binding domain and is associated with additional epigenetic changes such as recruitment of histone modifying enzymes [4]. Conversely, hypomethylation at promoters in general is associated with gene activation, although there is no simple inverse relationship.
DNA methylation is considered a favorable therapeutic target, as it can be reversible; however global alteration of methylation lacks specificity. Ten-eleven translocation 1 (TET1) enzyme and its role in DNA demethylation was identified in 2009. TET enzymes convert 5meC to 5-hydroxymethylcytosine (5hmC) (i.e., DNA hydroxymethylation) facilitating reactivation of gene transcription [5]. Given its recent discovery, not much is known regarding TET protein involvement, nor about hydroxymethylation, in fibrosis. The relationship between cytosine methylation and gene function is complex; methylation at non-CpG sites is now known to play a role in gene and enhancer function in numerous cell types [6].
Histone Modifications
Histone proteins are critical in chromatin structure and packaging. Lysine acetylation by histone acetyltransferases (HATs) reduces histone charge, loosening its electrostatic attraction to DNA and locally opening chromatin structure allowing for activation of gene transcription. P300, a histone acetylase and transcriptional coactivator, illustrates this concept. Activated Smad2/3 and p300 hyperacetylation of histone H4 at the collagen type 1 alpha-2 (COL1A2) locus activates TGFβ-induced collagen gene expression in myofibroblasts. Peroxisome proliferator-activated receptor γ (PPARγ) inhibits the TGFβ-induced recruitment of p300 to COL1A2 promoter [7]. Lysine acetylation can also create docking sites for bromodomains, where transcription-associated “reader” proteins can bind, creating multiprotein complexes that regulate transcription programs [8].
Deacetylation by histone deacetylases (HDACs) is associated with repression. HDAC4 has been described to most efficiently abrogate SMA expression and prevent TFGβ1-mediated morphological changes seen in myofibroblast differentiation in cultured human fibroblasts. HDAC4 also upregulates expression of the TGFβ1 signaling pathway repressors 5′-TG-3′-Interacting Factor (TGIF) and TGIF2 homeoproteins [9]. Histone hypoacetylation has been associated with profibrotic effects in pulmonary fibrosis [10] and systemic sclerosis [11, 12].
Histone methylation by specific histone methyltransferases (HMTs) can cause activation or repression of gene transcription. The most well-described histone methylations are histone H3 lysine 4 trimethylation (H3K4me3), an activator, and histone H3 lysine 9 di- and trimethylation (H3K9me2/3) and histone H3 lysine 27 trimethylation (H3K27me3) which are transcription repressors. Methylated histones function as a docking site for chromatin modifier proteins that facilitate downstream effects. For example, heterochromatin protein 1 (HP1) recruits additional HMT, DNMTs, and HDAC-containing complexes [13]. Methyl groups can be removed by histone lysine demethylases (KDMs). Exploration of KDM involvement in fibrosis is limited but have been demonstrated in liver HSCs activation to fibrotic myofibroblasts [14] and systemic sclerosis [15].
There is evidence to suggest that DNA methylation and histone modifications are closely interconnected in their regulation and promotion of gene silencing, which are also dependent on the physiologic environment and cell type [16]. The relationship between histone modification and DNA methylation seems to be bi-directional with each able to affect the other. This link may be facilitated by methyl-binding proteins that can recruit histone deacetylases to methylated areas [4].
Non-coding RNAs
A bi-directional relationship has also been described between non-coding RNAs (ncRNA) and DNA methylation. ncRNA are transcribed from DNA but not translated into proteins. Rather they are functional molecules that regulate gene expression at the level of gene transcription and post-transcriptional modifications. ncRNAs were recently reviewed by Beermann et al in 2016 [17].
MicroRNAs (miRs) are the most well described ncRNA involved in epigenetic regulation which bind to 3′ UTR of target genes and block translation of the target gene, leading to breakdown of target mRNA and/or blocking of protein translation. Abnormal expression of miRs is associated with disease processes including calcific aortic valve disease [18], cardiac fibrosis [19, 20], liver fibrosis [4, 21–24], lung fibrosis [25–30], renal fibrosis [31–33] and systemic sclerosis [34–43]. Multiple miRs may simultaneously and synergistically affect complex cellular processes such as EMT and these miRs may play different roles in varying organs or developmental stages [44]. The influence of different miRs on the development of fibrosis in various organs was reviewed by O’Reilly in 2016 [45]. Given their implication in the pathogenesis of various diseases, they have become a target for promising therapeutic interventions. Long non-coding RNAs (lncRNAs) are longer than 200 nucleotides and likewise influence gene transcription and protein translation in developmental and disease processes [17].
Taken all together, seemingly separate epigenetic mechanisms construct complex epigenetic regulatory networks which shape whole gene expression profiles, and the disruption of these circuits interrupts normal functions and causes disease.
In this review, we highlight epigenetic mechanisms that have been elucidated to shape myofibroblast behavior in different organs and disease processes related to fibrosis. It can be difficult to discriminate myofibroblasts from their precursor cells, or even to measure myofibroblast de-differentiation into a more quiescent state. Thus, we assume that studies referring to fibroblasts that play a role in fibrosis without specifically mentioning myofibroblasts are essentially describing a myofibroblast phenotype. As hundreds of individual genes can be epigenetically altered in myofibroblasts, where relevant we have referred to recent reviews on epigenetically-regulated genes by organ, and here we focus on mechanisms. Table 1 is a collection of epigenetic regulators and the mechanism by which they influence myofibroblast phenotype in different organ-specific fibrotic diseases. This table and review is by no means a complete compilation of all epigenetic mechanisms involved but we strive to present a starting point for readers to unravel overlapping and organ/disease-specific epigenetic drivers of myofibroblast phenotype in the context of fibrosis. (Table 1)
Table 1.
Organ | Disease | Epigenetic Regulator(s) | Effect | Mechanism and Findings | References | |
---|---|---|---|---|---|---|
Heart | Fibrosis | DNA methylation Hydroxymethylation |
EndoMT →
myofibroblast ↓EndoMT |
[TGFβ1, hypoxia → aberrant
methylation RASAL1 gene promoter ⦰ RASAL1] → ↑ Ras-GTP
activity BMP7 → TET3 hydroxymethylation of induced RASAL1 promoter methylation → RASAL1 |
[46, 47] | |
Fibrosis | DNA methylation | Overactive fibrotic phenotype | Hypoxia → human cardiac fibroblasts express hypoxia-inducible factor (HIF-1α) → → DNA methylation → pro-fibrotic genes | [48] | ||
Fibrosis | DNA methylation | Fibroblast activation | [↑ → DNA methylation ⦰ Ras association domain family 1 isoform A (RASSF1a)] → ERK1/2 | [110] | ||
Myocardial infarction and myocardial rupture | DNA methylation | Proliferation and differentiation (myo)fibroblast-like cells → ECM degradation and fibrogenesis | Deficient NEIL3 → dysregulation methylation in cardiac epigenome | [58] | ||
Fibrosis and hypertrophy | DNA methylation inhibitor 5-azacytidine ( ) | ⦰ fibroblast activation → ↓myocyte size |
⦰ myofibroblast differentiation → ↓
TGFβ1-induced collagen I/III and SMA mRNA expression and ↓myocyte size → ↑MYBP-C3 gene expression in myofibroblasts |
[99, 100] | ||
Fibrosis | HDAC6 | Fibroblast proliferation | ⦰ RASSF1a→ ERK1/2 | [50] | ||
Fibrosis | Class I HDACs inhibitors | ⦰ Angiotensin II dependent cardiac fibrosis,
fibroblast activation and differentiation ⦰Differentiation bone marrow-derived fibrocytes |
[ → p15, p57 ⦰ cyclin-dependent kinase] ⦰
retinoblastoma protein phosphorylation ⦰ G1-to-S phase transition →
↓ECM-producing cells ⦰ ERK1/2 |
[51] | ||
Congestive heart failure after MI | HDAC1 inhibitor ( ) | [Reversal of cardiac myofibroblast phenotype to fibroblast and induction cell cycle arrest/apoptosis] → ↓ ECM production, ↓interstitial fibrosis, ↓scar size |
→ 1. Reversal of myofibroblast phenotype (↓SMA, ↓collagen III, ↓MMP-2) 2. ↑E-cadherin and surface translocation β-catenin → CD 90+ fibroblast transformation to epithelial-like phenotype 3. ↓phosphorylation Akt (↓EMT/EndoMT) in CD 90+ fibroblast 4. Induction cell cycle arrest/apoptosis (fibroblasts in vivo) → ↓/reverse fibrosis, ↓ ECM production |
[53] | ||
Systolic Heart Failure (HF) | HDAC inhibitor ( ) | ⦰ Cardiac fibroblast proliferation and migration | on isolated primary cardiac fibroblasts from HF rats → ↓TGFβ1, ↓angiotensin II type I receptor (AT1R), ↓migration and proliferation cardiac fibroblasts (↓↓ in presence of TGFβ1) | [54] | ||
Atrial fibrosis and arrhythmias in cardiac hypertrophy | (non-selective HDAC inhibitor) | Reverses atrial fibrosis | HDAC → cardiac hypertrophy → reverses atrial fibrosis, connexin 40 remodeling and ↓ atria arrhythmia vulnerability independent of angiotensin II |
[55] | ||
Myocardial disease and failure | miR21 | → fibroblast survival and fibrosis | ↑ in cardiac fibroblasts of failing heart ⦰ sprout homologue 1
(Spry1) → ERK-MAP kinase pathway → fibroblast survival and growth
factor secretion → interstitial fibrosis and cardiac
hypertrophy [ antagomir in mouse pressure-overload induced disease ⦰ ERK-MAP kinase activity ⦰ interstitial fibrosis] → ↑cardiac function |
[111] | ||
Myocardial infarction (MI) and fibrosis | family | ⦰ mRNAs involved in ECM production and fibrosis | MI → ↓ in regions adjacent to infarct → ↑collagen expression
(COL1A1, COL1A3, COL3A1 and fibrillin 1-FBN1) ↑ in fibroblasts → ↓collagen |
[19] | ||
MI | miR145 | Cardiac fibroblasts → myofibroblast phenotype and mature collagen → scar healing and contracture | Coronary occlusion and hypoxia → ↓
then ↑ → MTD with ↑SMA |
[112] | ||
Fibrosis and hypertrophy | miR199b | Pro-fibrotic | [⦰ ] → normalized Dyrk1a expression → ↓
nuclear NFAT activity → inhibition and reversal of hypertrophy and fibrosis in
mouse heart failure model Dyrk1a ⦰ [cardiac stress → calcineurin/NFAT signaling → cardiac hypertrophy/remodeling and failure] + NFAT ⦰ Dyrk1a |
[20] | ||
Calcific Aortic Valve Disease | miR-486 | Aortic valve interstitial cells → myofibroblast phenotype | ↑ → ↓ phosphatase and tensin homolog (PTEN) expression, ↑ phosphorylated protein kinase B (AKT) phosphorylation in AVICs → ↑ SMA expression | [18] | ||
Pressure overload-induced cardiac fibrosis | , and | ↓ → fibrotic phenotype | on enhancer regions → ↓ → ↑connective tissue growth factor expression, ↑ fibrosis, ↑ left atrium diameter (↓ diastolic function) | [60] | ||
Liver | Fibrosis | DNA hypermethylation | HSC → myofibroblast phenotype → fibrosis | Methylation by →loss Septin 9 expression | [64] | |
Fibrosis | DNA methylation | Methylation CTGF gene promoter ⦰ [HSCs → myofibroblast] | [↑ methylation at connective tissue growth factor (CTGF) gene promoter ⦰ CTGF] ⦰ HSCs phenotypic changes into myofibroblast (+SMA) | [65] | ||
Fibrosis | DNA methylation | HSC → myofibroblast phenotype → fibrosis | [DNA methylation Smad7 gene ⦰ Smad7 repression]→ phosphorylation and activation TGFβ-mediated Smad2/3 pathways | [66] | ||
Fibrosis | DNA hypermethylation | HSC → myofibroblast phenotype → fibrosis | methylation of phosphatase and tension homolog deleted on chromosome 10 (PTEN) gene promoter → PI3K/AKT and ERK pathways | [67] | ||
Fibrosis | DNA methylation and | Sustain fibroblast activation → fibrosis | [ and hypermethylation Patched (PTCH1) ⦰ PTCH1] → perpetuation fibroblast activation and fibrosis | [68] | ||
Fibrosis and wound healing | DNA methylation and |
Loss of IϰBα and PPARγ expression → proinflammatory and profibrotic myofibroblast characteristics | [ interacts with IϰBa promoter via methyl-CpG sites + CBF-1
bound to NF-kB (kB2)/CBF-1 site] → recruited to IϰBα promoter → histone modification → transcriptional repression |
[63] | ||
Fibrosis | DNA methylation, , and Methylation histone 3 lysine 27 ( ) and histone 3 lysine 9 ( ) |
HSC → myofibroblast phenotype → fibrosis |
downregulated → translation → binds to PPARγ → → HP1α recruitment → chromatin condensation
⦰ PPARγ → EZH2 expression → → binding site for Polycomb repressor 1 complex → chromatin condensation ⦰PPARγ |
[4] | ||
Fibrosis | HDAC4 | HSC transdifferentiation and ⦰ collagen degradation | [ → ↓histone acetylation ⦰ IL-1 induced MMP promoter
activity and MMP9 expression] → HSC activation IL-1 → MMP 9 → collagen IV degradation |
[69] | ||
Fibrosis | HDAC7 | → pro-fibrotic HSCs | [ ⦰ HGF in HSCs] → fibrosis CYLD removes from HGF promoter ⦰ fibrosis |
[70] | ||
Alcohol-induced liver disease and fibrosis | (H3K4 methyltransferase) | →pro-fibrotic HSC transition with ↑ECM production | Ethanol → recruitment to elastin gene promoter→ → ↑ elastin expression in transitioning
HSCs ↑ → ↑elastin/collagens in alcoholic liver disease explants Ethanol → ↑profibrogenic/ECM gene expression |
[71] | ||
Fibrosis | Absent, small or homeotic disc 1 ( ) – histone methyltransferase of lysine 4 at histone H3 (H3K4) and | HSC → myofibroblast phenotype → fibrosis |
binds to regulatory regions of SMA, collagen I, TIMP1, TGF-β1 in
activated HSCs → H3K4 methylation and gene activation → expression |
[113] | ||
Fibrosis | HAT and coactivator | → HSC activation and ↑collagen | TGF-β → /CBP → histone H3 hyperacetylation at VDR/SMAD COL1A1 regulatory
region→ HSC activation Calcipotriol (synthetic VDR agonist) ⦰ VDR/SMAD interaction →↓collagen deposition and fibrotic gene expression |
[72] | ||
Fibrosis | Jumonji domain-containing protein 1A ( ) – H3K9 demethylase | Modulates HSCs activation → | Small interfering RNA → knockdown in HSCs→ H3K9me2 PPARγ gene promoter → ↓PPAR γ mRNA and protein → ↑SMA/COL1A expression, strengthened collagen production, enhanced necrosis | [14] | ||
Fibrosis | Histone methyltransferase inhibitor 3-deazaneplanocin A ( ) | ⦰ fibrosis progression in vivo despite sustained liver damage | Myofibroblast targeting antibody-liposome- → ↓EZH2 → ↓ H3K27me, H3K4me3, H3K9me3, and H3K36me3 | [98] | ||
Fibrosis | (non-selective HDAC inhibitor) | ⦰ EMT and fibrosis |
⦰ TGF-β1-induced EMT] ⦰ fibrosis ⦰ p300 binding to SMAD3 ⦰ activation of type I collagen promoter ⦰ type I collagen deposition → Friend leukemia virus integration 1 ⦰ type I collagen gene |
[101] | ||
Fibrosis |
|
→ HSC activation and proliferation | TGF-β1-treated HSCs and CCl4-induced rat fibrosis
→ ↑ ⦰ [SMAD7 ⦰
TGF-β1] ⦰ ⦰ HSC proliferation, apoptosis resistance |
[24] | ||
Fibrosis | miR29 | ↓ → HSCs → fibrosis | ↓ in murine carbon tetrachloride-induced hepatic fibrosis and human fibrotic
livers TGF-β, NF-kB, lipopolysaccharide (LPS) → ↓ in HSCs → ↑ extracellular matrix genes ↑ in HSCs → ↓ collagen expression |
[21] | ||
Fibrosis | miR33a | HSC activation → fibrosis | TGF-β1 → [↑ ⦰ P13K/Akt pathway, PPAR-α mRNA/protein] → HSC activation and extracellular matrix production | [23] | ||
Fibrosis | miR133a | ↓collagen synthesis in HSCs | TGF-β → ↓ in HSCs during fibrogenesis ↑ ↓collagen expression |
[22] | ||
Fibrosis | (BRD4 inhibitor) | ↓ [cytokine-induced activation of HSCs → myofibroblasts] | JQ1 ⦰ TGFβ-mediated activation
pro-fibrotic gene expression JQ1 reversed pre-existing liver fibrosis in CCl4 mouse model |
[114] | ||
Lung | Fibrosis | DNA methylation and | HELF cells → myofibroblast → fibrosis | → methylation of tuberous sclerosis TSC1, TSC2 promoter regions ⦰ TSC1, TSC2 protein expression | [73] | |
IPF | DNA methylation (non-selective HDAC inhibitor) |
→fibrogenic phenotype | Thy-1 ⦰ MTD from fibroblast DNA hypermethylation at Thy-1 promoter → fibrogenic myofibroblast → fibrosis DNA methyltransferase inhibitors → restoration Thy-1 expression in Thy-1(−) fibroblasts → [↓H3K27me3 → demethylation at Thy-1 promoter, restoration Thy-1 expression] and ↑histone acetylation (H4) and ↑H3K4me3 |
[76] [78] |
||
IPF | Methylation histone 3 lysine 9 ( ) and histone 3 lysine 27 ( ) | Promotes fibrogenic phenotype, increases collagen production | [G9a- and EZH2-binding → and ↔ recruitment and HDAC-containing complexes to cyclooxygenase-2 (COX-2) promoter] ⦰ COX-2 → loss prostaglandin E2 | [16] | ||
IPF | HDAC4 | Normal human lung fibroblasts → myofibroblasts | [ ⦰ protein phosphatase-mediated dephosphorylation AKT] → TGF-β1 → phosphorylated AKT | [81] | ||
IPF | Elevated | Increase fibroblast proliferation, fibroblast-to-myofibroblast differentiation, apoptosis-resistant myofibroblast phenotype | Upregulation of myofibroblasts in fibroblastic foci → AKT signaling pathways and TGF-β1 induced differentiation of normal lung fibroblasts into myofibroblasts (pan-HDAC inhibitor) on primary fibroblast → ↓expression of ECM synthesis, ↓proliferation and cell survival, ↓HDAC7 genes, and [endoplasmic reticulum stress → apoptosis] (class I HDAC inhibitor) → partial attenuation profibrotic and apoptosis-resistant phenotype of IPF fibroblasts |
[80] | ||
IPF | Histone acetylation and | ↓ apoptosis resistance in fibroblasts | ↓histone acetylation, ↑ at Fas promoter → ↓Fas → ↓Fas-mediated apoptosis resistant fibroblasts | [79] | ||
IPF | Suberoylanilide hydroxamic acid ( , HDAC inhibitor) | ↑myofibroblast apoptosis susceptibility | → ↑histone modifications (acetylation H3K9, H3, H4 and methylation H3K9Me3), ↓DNMT1/3a mRNA →↑pro-apoptotic Bak with promoter hypomethylation, ↓anti-apoptotic Bcl-xL→ (myo)fibroblast apoptosis | [82] | ||
IPF | (selective Class I HDAC inhibitor) | ⦰ fibroblast proliferation and collagen production | → ↑histone 3 acetylation, ↑cell-cycle inhibitor p21waf1, ↓ SMA and interstitial collagen expression | [83] | ||
IPF | Histone hypoacetylation | Uncontrolled fibroblast proliferation | [↓ histone acetyltransferases→↓histone H3 and H4 acetylation] and ↑ transcription of HDAC-containing corepressor complexes to COX-2 promoter → ↓ transcription factor binding → ↓ COX-2→ ↓ prostaglandin E2 (antifibrotic mediator) | [10] | ||
IPF | (KMT1C) | Activation fibroblasts → fibrosis |
→ methylates H3K9 ↑ , heterochromatin protein 1 (HP1) to gamma interferon-inducible protein of 10 kDa (IP-10) promoter in IPF lung fibroblasts → repressive chromatin → ↓anti-fibrotic (IP-10) Small molecule inhibitor → ↓hypermethylation and ↑HP1/CoREST binding to promoter → restored expression IP-10 |
[13] | ||
Fibrosis | and | Pro-fibrotic myofibroblast phenotype | TGFβ and PDGF stimulation lung fibroblasts
→ → histone acetylation → binds → enhanced transcription → SMA induction of fibroblasts inhibitor → prevent fibrosis after intratracheal bleomycin, [↓ PDGF- and TGFβ-stimulated IL-6 secretion, collagen and SMA expression, and proliferation/contraction cultured lung fibroblasts] |
[8, 84] | ||
IPF | cluster and DMNT1 | Fibrotic gene expression → pro-fibrotic phenotype | ↓ → ↑DNMT1 → ↑DNA methylation promoter | [29] | ||
IPF | Increased expression primarily in myofibroblasts | Promotes fibrosis | TGF-β1→ expression → Smad7 inhibition, Spry1 expression →
TGF-β1 induced fibrogenic activation of pulmonary
fibroblasts bFGF → expression in human primary fibroblasts |
[25] | ||
Fibrosis |
|
↓ → fibrotic phenotype with ECM deposition and remodeling | TGFβ →↓ → ↑ expression profibrotic target genes (collagens,
ECM-associated, remodeling, laminins, integrins), ↑ severity of fibrosis targets and negatively regulates SMAD3 in TGFβ pathway and CTGF expression |
[27] [28] |
||
IPF | (miR) | ↓ → EMT, ↑ collagen deposition | TGF-β1 → SMAD3 binding to promoter →↓ → ↑ mesenchymal markers N-cadherin, vimentin, SMA, HMGA2 | [26] | ||
IPF | miR199a-5p | Fibroblast → myofibroblast → fibrosis | ↑ in myofibroblasts from injured mouse lung and fibroblastic
foci ectopic → pulmonary fibroblasts proliferation, migration, invasion, and differentiation into myofibroblasts TGFβ → ↑ → TGFβ-induced ↓caveolin-1 (CAV1) |
[30] | ||
Kidney | Fibrosis | DNA hypermethylation | Fibroblast → myofibroblast → fibrosis | → hypermethylation RASAL1 → irreversible transcriptional repression | [85] | |
Fibrosis and renal anemia | DNA methylation | Demethylating 5-azacytidine → myofibroblast re-differentiation into pericytes | Renal erythropoietin-producing cells (REPCs) in fibrotic
kidneys Kidney pericytes can differentiate into collagen-producing myofibroblasts Fibroblast-like FOXD1+ progenitor-derived kidney pericytes produce erythropoietin [FOXD1+ pericytes differentiate into myofibroblasts → hypermethylation erythropoietin] ⦰ erythropoietin production Demethylating 5-azacytidine on myofibroblasts 1. → erythropoietin ⦰ anemia 2. re-differentiate myofibroblasts into pericytes |
[86] | ||
Interstitial fibrosis secondary to obstructive nephropathy | (non-selective HDAC inhibitor) | Prevent activation and accumulation fibroblasts | ⦰ SMA and fibronectin expression, phosphorylated STAT3 and accumulation activated renal interstitial fibroblasts after obstruction | [87] | ||
Interstitial fibrosis secondary to obstructive nephropathy | Class III HDAC sirtuin ( and ) | Renal fibroblast activation, proliferation and fibrogenesis |
expressed in cultured renal interstitial fibroblasts ( inhibitor), ( inhibitor), ( inhibitor), small interfering RNA → ↓fibroblast activation markers (SMA, fibronectin, collagen I) and proliferation markers (proliferating cell nuclear antigen, cyclin D1/E → dephosphorylation EGFR, PDGFRβ, STAT3 |
[88] | ||
Fibrosis secondary to obstructive nephropathy | miR29 | ⦰ TGF-β1/SMAD3 mediated
fibrosis ⦰ [TGFβ → ADAM12/19] mediated fibrosis |
TGF-β1 → SMAD3 binding to promoter →↓ in cultured fibroblasts → ↑ collagen I/III expression gene delivery before or after obstructive nephropathy ⦰ fibrosis ⦰ [TGFβ-mediated upregulation ADAM12/19 → epithelial to fibroblast phenotype differentiation with ↑Col1a/Col3a] family has no effect on SMA/MMP1 |
[32] [33] |
||
Fibrosis in diabetic nephropathy | miR-129 | ⦰EMT | [↑ ⦰ ZEB1/2] ⦰ TGFβ-mediated downregulation
E-cadherin in proximal tubular cells ↓ → tubulointerstitial fibrosis and low GFR |
[31] | ||
Fibrosis in diabetic nephropathy | HAT | Tubulointerstitial fibrosis | High glucose → (requires myocardin-related transcription factor MRTF-A a potent
pro-fibrotic co-factor for serum response factor) → ECM genes → type 1
collagen ⦰ MRTF-A → disappearance acetylated H3k18/K27 and trimethylated H3K4 MRTF-A recruits and WD repeat-containing protein 5 (key H3K4 methyltransferase complex) to collage promoters → transcription |
[115] | ||
Other | Systemic Sclerosis | DNA hypermethylation | Persistent fibroblast activation | [Ddickkopf-1 (DKK1) and secreted frizzled-related protein-1 (SFRP) hypermethylation ⦰ DKK1/SFRP] → canonical Wnt signaling | [116] | |
Systemic Sclerosis | DNA hypermethylation, histone hypoacetylation, and histone deacetylation | Fibroblast activation→ profibrotic effects | [Friend-leukemia integration factor 1 (FLI1)
hypermethylation and H3/H4 hypoacetylation ⦰ FLI1] →
↑collagen type 1 gene expression 2-deoxy-5-azaC ( ) and Trichostatin A ( , non-selective HDAC inhibitor) → ↓ type 1 collagen expression |
[11] | ||
Systemic Sclerosis | DNA hypermethylation and histone hypoacetylation | Sensitizes fibroblasts to profibrotic stimuli | Kruppel-like factor 5 (KLF5) hypermethylation and histone
H3/H4 hypoacetylation ⦰ KLF5 Epigenetic downregulation KLF5 and FLI1 synergistically →↑ connective tissue growth factor |
[12] | ||
Systemic Sclerosis | (non-selective HDAC inhibitor) | Inhibits SSc fibroblast
phenotype ⦰fibroblast proliferation and extracellular matrix accumulation |
[ ⦰ Wnt inhibitor factor 1 (WIF-1) loss] ⦰ → → treatment of SSc skin fibroblasts →↓ collagen type 1 alpha-1 (COL1A1) and fibronectin →↓ collagen protein → ↑cell-cycle inhibitor p21 |
[117] [118] |
||
Systemic Sclerosis | (Class II HDAC) | ⦰ →↓ECM |
⦰ →↓constitutive and cytokine-induced production type I/III
collagen, no ↑ profibrotic molecules (ICAM-1, CTGF) TSA, in addition to anti-fibrotic effects, also →↑ ICAM-1/CTGF, ↑ more specific to SSc → silencing more effective and safer than |
[119] | ||
Systemic Sclerosis | and |
→ SSc fibroblasts with increased ECM production → ↓ collagen |
[↑ → hypermethylation promoter ⦰ ] → STAT-6-mediated ECM induction → ↓ IL-13 mediated collagen induction |
[45] | ||
Systemic Sclerosis | HAT and transcriptional coactivator | Persistence and progression of fibrotic response by SSc fibroblasts | TGFβ in presence of → TGFβ-induced collagen gene → ↑ collagen
production TGFβ → early-immediate TF (Egr-1) expression → transcription COL1A2, [ → COL1A2 promoter histone acetylation → SMAD2/3 transcription activation] |
[120] [7] |
||
Systemic Sclerosis | Class III histone deacetylase sirtuin 1 (SIRT1) | Fibroblast enhanced release of
collagen Antifibrotic effects |
SIRT1 →TGFβ/Smad signaling
→release of collagen Conflicting outcome: SIRT1 →↓ →↓ collagen |
[121] [122] |
||
Systemic Sclerosis | Mitochondrial deacetylase sirtuin 3 ( ) | Mitigates activated SSc fibroblast phenotype |
⦰ intracellular TGFβ signaling and fibrotic responses reduced in SSc skin and fibroblasts Downregulation SIRT3 → mRNA profibrotic genes In vivo, (pharmacologic agonist) →↓ bleomycin-induced skin and lung fibrosis |
[123] | ||
Systemic Sclerosis |
3-deazaneplanocin A ( ) – H3K27me3 inhibitor |
activation of fibroblasts and myofibroblast differentiation | [ ⦰ ] → release of collagen from fibroblasts, ↑
profibrotic transcription factor fos-related antigen 2 (FRA-2) → ↑FRA-2→ TIMP→ fibroblast-to-myofibroblast differentiation |
[124] [125] |
||
Systemic Sclerosis | H3K27me3 demethylase Jumonji domain containing protein 3 ( ) | Fibrosis phenotype | ⦰ →↑ , ↓FRA-2, ↓contractile fibers, ↓SMA,
↓collagen secretion, ↓myofibroblast
differentiation TGFβ → expression ⦰ →↓ fibrosis in bleomycin-, Topo- and TBRact-induced experimental models |
[15] | ||
Systemic Sclerosis | miR7 | ↓collagen and anti-fibrotic phenotype | ↑ thrombospondin-2 (TSP-2) protein in SSc dermal
fibroblasts → ↑ ⦰ TSP-2 mRNA ⦰ collagen expression |
[34] | ||
Systemic Sclerosis | and | Anti-fibrotic phenotype with collagen degradation and ↓ ECM/collagen production |
⦰ [TGFβ activated kinase 1 binding protein 1
(TAB-1) → TIMP-1 production → ECM
deposition] ↓ → TGFβ, PDGF ⦰ → matrix metalloproteinase-1 (MMP-1) → collagen degradation targets COL3A1 post-transcription →↓ collagen production SSc skin/fibroblasts and TGFβ-stimulated fibroblasts → ↓ →cCOL1A1 mRNA |
[35, 36] [37] |
||
Systemic Sclerosis | miR129-5p | ↓collagen and ↓fibrotic characteristics | ↓ in SSc fibroblasts ↑ ↓connective tissue growth factor, ↓α1(I) collagen |
[38] | ||
Systemic Sclerosis | and DNA methylation | ↓ → fibrosis with ↑ collagen | DNA methylation →↓ in SSc dermal fibroblasts → integrin 3 → collagen type 1 overexpression | [39] | ||
Systemic Sclerosis | miR196a | ↓ → fibrosis with ↑ collagen | TGFβ-stimulated fibroblasts and Ssc fibroblasts
→↓ ↓ →↑ collagen type 1 expression |
[40] | ||
Systemic Sclerosis | miR206 | ↓ → fibrosis | SSc skin → ↓ regulates 15 genes related to SSc pathogenesis: TGFβ1, TGFβ2, SMAD5, FGF2, FGF5, FGFR3, IL1β, IL1R AP (receptor accessory protein), IL21, integrinα2, cyclinT2, FGβ (fibrinogen β chain), COL6α3, COL29α2 and CD28 |
[41] | ||
Systemic Sclerosis |
miR145 |
Fibrosis phenotype | Fibroblasts exposed to TGFβ → 1. ↑ →↓SMAD7 mRNA 2. ↑miR145 →↓SMAD3 mRNA Conflicting outcome: ↓miR145 and ↑ SMAD3 mRNA in SSc skin biopsies and SSc fibroblasts |
[37] | ||
Systemic Sclerosis | miR155 | → collagen synthesis and skin, lung, and systemic fibrosis | ↑ in fibrotic skin from localized scleroderma and
SSc ⦰ in primary skin fibroblasts→ 1. ⦰collagen synthesis 2. ⦰Wnt/β-catenin and Akt pro-fibrotic pathways targets 1. casein kinase 1α (CK1α) ⦰β-catenin pathway 2. Src homology 2-containing inositol phosphatase-1 (SHIP-1) ⦰ Akt signal pathways topical antagocmiR-155 treats bleomycin-induced skin fibrosis in mice ((↓β-catenin pathway and ↓phosphorylated Akt staining in fibroblasts) (−/−) mice resistant to bleomycin-induced skin fibrosis (↓β-catenin pathway, ↓phosphorylated Akt) (−/−) protected from bleomycin-induced pulmonary fibrosis |
[42] [43] |
||
Systemic Sclerosis | lncRNA | fibrosis with ↑ collagen | TGFβ →↑ →stabilizes COL1A1 mRNA → ↑type I collagen | [126] | ||
Eye | Pterygium | family | → EMT | and → EMT, ↑ fibronectin (FN1) | [91, 92] |
By organ-specific fibrotic diseases, epigenetic regulators, their effect on myofibroblast phenotype, and mechanism of action. Epigenetic regulators in and illicit an antifibrotic versus profibrotic effect respectively.
→ = leads to or stimulates, ⦰ = inhibits, ↑= upregulates, ↓ = downregulates, [ ] groups events in a mechanism
IV. Epigenetic Regulation of Myofibroblast Phenotypes in Organ Fibrosis
Heart
After injury or stress, initial reparative cardiac remodeling can adversely evolve into fibrosis, replacing normal heart tissue with non-functional fibrotic myocardium. This stiff tissue causes ventricular dysfunction and essentially heart failure. As heart failure is a leading public health problem with very high mortality rates, pursuing therapies directed at these fibrotic alterations is vital. Activated cardiac fibroblasts, fibroblast-like cells from endothelial-to-mesenchymal transition, and possibly circulating bone-marrow-derived fibrocytes transdifferentiate into myofibroblasts and are the principal drivers of cardiac fibrosis. Epigenetic changes including DNA methylation [46–49] and hydroxymethylation [46], histone modifications [50], and miRs [20] have been implicated in cardiac fibrosis.
During activation of fibroblasts, histone deacetylases (HDACs) are up-regulated and thereby likely take part in cardiac fibrosis; however, the exact molecular mechanisms linking specific histone modifications and fibrosis remain unknown. HDAC inhibitors have been used to infer mechanisms such as blockage of cardiac fibroblast proliferation and migration, induction of genes that suppress fibroblast ECM production, repression of pro-inflammatory markers, and endoMT prevention [51–55]. HDAC inhibitors also block collagen synthesis in isolated cardiac fibroblasts and fibrotic changes diminished in pressure-overload cardiac hypertrophy [56]. Lysine methyltransferase DOT1L belongs to the KMT4 family and seems to be involved in cardiac fibrosis and hypertrophy though it is unclear whether KMT directly orchestrates activation of fibroblasts in cardiac fibrosis [57].
After myocardial infarction (MI), a repair process engages fibroblasts to proliferate, differentiate, and deposit ECM for scar formation and stabilization. An imbalance in the breakdown and deposition of ECM can cause poor repair, aneurysm formation, infarction expansion, and even detrimental myocardial rupture. Myofibroblasts are the primary source of ECM and its modulators after an MI. Fibroblasts, endothelial cells, epithelial cells, smooth muscle cells, and pericytes are considered cardiac myofibroblast precursors. Regulation of precursor cell morphology, growth, proliferation, and migration to infarcted areas are crucial to appropriate healing [57].
Calcific aortic valve disease (CAVD) is a common cardiovascular disease that involves fibrosis and calcification of aortic valve leaflets for which there is no pharmacological treatment [58]. Aortic valve interstitial cells (AVICs) preserve the structure and function of the aortic valve. In healthy human valves, AVICs mainly take on a fibroblastic phenotype and produce ECM to retain normal valvular structure. In contrast, AVICs adopt a myofibroblastic phenotype and are abundant in calcified aortic valves and their augmented proliferative activity and overproduction of ECM protein cause valve thickening and fibrosis [59]. miRs have been implicated as an epigenetic regulator of AVIC phenotypic transition [18].
Examples of multiple epigenetic mechanisms acting together to affect myofibroblast phenotype can be illustrated in cardiac fibrosis. Hydroxymethlyation by TET3 induces DNA methylation which downregulates endoMT inhibiting fibrosis [46, 47]. In pressure overload-induced cardiac fibrosis, histone deacetylases HDAC1 and HDAC2 counteract miR133a antifibrotic signaling [60]. Additional epigenetic modifications involved in cardiac fibrosis and therefore, myofibroblast phenotypes can be found in recent reviews [56, 57].
Liver
Repetitive liver injury and almost all chronic liver diseases cause liver fibrosis. Progressive fibrosis leads to liver failure and in some circumstances, even hepatocellular carcinoma [1]. Liver fibrosis was previously regarded as a process that was beyond repair but compelling evidence now indicates that the removal of inciting triggers and apoptosis of activated hepatic stellate cells (HSCs) can reduce fibrosis. In the liver, myofibroblasts can originate from HSCs which drives the pathogenesis of liver fibrosis. Or, as suggested by Karin et al, the origin of myofibroblasts changes based on whether injury originates in the hepatic lobule such as in chronic hepatitis versus portal areas in biliary diseases. Activated HSCs are considered the “myofibroblast for hepatocytes” and activated portal myo(fibroblasts) the “myofibroblast for cholangiocytes” [61]. Recently, the epigenetic regulation of HSC activation in liver fibrosis was reviewed [1]. In 2016, Nwosu et al summarized DNA methylation and histone modifications described in liver fibroblasts and anti-fibrotic therapies that target these epigenetic mechanisms [62].
The epigenetic control of myofibroblast transdifferentiation was validated with the application of 5-aza-2′-deoxycytidine (5-azadC), a DNA methylation inhibitor, to cultured HSCs which suppresses MTD features [63]. Subsequently, DNA methylation of genes engaged in myofibroblast activation and fibrogenesis have been identified [62, 64–68]. Hepatic MTD seems to be epigenetically regulated by its DNA methylome through direct histone methylation and changes in chromatin structure [4, 63].
PPARγ, a master antifibrogenic gene, negatively regulates myofibroblast transdifferentiation. MiR132, MeCP2, and EZH2, a lysine methyltransferase, guide an epigenetic relay pathway that transcriptionally represses PPARγ. MeCP2 is absent in normal human and rat livers but strongly upregulated during culture-induced transdifferentiation of HSCs and diseased livers suggesting that MeCP2 is a key regulator of the myofibroblast phenotype [63]. Furthermore, MeCP2 leads to hypermethylation of Patched (PTCH1) whose repression is critical to maintain HSC activation and hepatic fibrosis [68]. A histone H3K9 demethylase Jumonji domain-containing protein 1A (JMJD1A) directs HSC activation through PPARγ expression [14]. DNA methylation also inhibits Smad7 granting TGFβ-mediated Smad2/3 phosphorylation activating fibrogenesis [66] and it governs IϰBα inflammatory effects. Attenuation of IϰBa is necessary to stimulate NF-κB activity that leads to MTD with expression of pro-inflammatory genes and myofibroblast resistance to apoptosis [63]. Hence, DNA methylation controls key inflammatory (IϰBα) and fibrogenic (PPARγ) transcriptional regulators of the myofibroblast phenotype.
Additionally, histone modifications are also operational during HSC activation and differentiation. Upregulated MMP9 causes collagen IV degradation and can therefore counter myofibroblast-stimulated ECM production. HDAC4 has been pinpointed as the culprit in the suppression of IL-1 induced MMP9/13 allowing for HSC transdifferentiation [69]. Hepatocyte growth factor (HGF) is involved in the fibrotic response to acute injury and cylindromatosis (CYLD) is an anti-fibrotic tumor-suppressor gene. HDAC7 inhibits HGF increasing HSCs susceptibility to fibrosis. CYLD removes HDAC7 from the HGF promoter preventing fibrosis [70]. Interestingly, ethanol can stimulate epigenetic modifications in hepatic stellate cells [71]. Excess alcohol consumption causes liver disease and fibrosis is a common feature of disease progression. Ethanol stimulates histone modifications changing chromatin structure and directly impacting HSC transdifferentiation with resultant ECM protein expression. Vitamin D receptor (VDR) ligands and histone modifications can also influence HSC transdifferentiation [72].
Lung
Idiopathic pulmonary fibrosis (IPF) is a progressive, eventually fatal disease with limited effective treatments. Fibrogenesis is driven in large part by the transdifferentiation of pulmonary fibroblasts into myofibroblasts [73]. Prevention of fibroblast transdifferentiation into myofibroblasts is an effective strategy to assuage pulmonary fibrosis. The epigenetic players in IPF have been reviewed [74]. A review of microRNAs that play a role in idiopathic pulmonary fibrosis was published in 2011 [75]. As the epigenetic modifications in IPF have been extensively reviewed, we will provide a limited overview of epigenetic regulators of myofibroblasts in lung fibrosis.
Altered DNA methylation in a large number of genes is associated with fibroblast activation and myofibroblast proliferation [73, 76], and global changes in DNA methylation are seen in IPF [77]. Starting in 2008 descriptions of epigenetic processes driving fibrosis in IPF began to appear, with Sanders et al demonstrating that DNA methylation at the Thy-1 promoter inhibited Thy-1 expression, stimulating MTD and myofibroblast apoptotic resistance [76]. Later in 2011, Sanders et al further demonstrated that histone acetylation affects Thy-1 expression and histone modifications can alter DNA methylation [78]. In 2013, Huang et al demonstrated that histone modifications led to the downregulation of FAS with resultant FAS-mediated apoptosis resistant fibroblasts in lung fibrosis [79]. In 2015, for the first time, Korfei et al demonstrated the involvement of specific histone deacetylases in IPF [80]. Since then, multiple studies have linked histone deacetylase enzymes to pathologic (myo)fibroblast behavior [81–83]. Bromodomain and extra-terminal domain-containing proteins (Brd) bind to acetylated histone residues triggering recruitment of components necessary for gene transcription. Brd2 and Brd4’s role in influencing lung fibroblast phenotype in fibrosis was first demonstrated in 2013 [8, 84].
Kidney
Renal fibrosis occurs when physiologic wound healing fails to cease and transitions into pathologic scarring and progressive organ failure. In renal fibrosis, the proliferation of activated (myo)fibroblasts in addition to increased deposition of ECM and increased SMA expression is considered the hallmark of fibrosis. DNA hypermethylation has been shown to be associated with fibroblast activation and proliferation of pro-fibrogenic myofibroblasts [85]. Remarkably, activated myofibroblasts can play a role in the development of anemia by stimulating epigenetic changes. By comparing two models of renal fibrosis, Chang et al found that fibroblast-like FOXD1+ pericytes differentiate into myofibroblasts and this transition is associated with decreased erythropoietin production and a dampened response by these cells to anemic stimuli. Methylation studies reveal hypermethylation of the erythropoietin gene and application of a demethylating agent on isolated myofibroblasts from these fibrotic kidneys restores erythropoietin expression at baseline and in response to hypoxia [86]. HDAC [87, 88] and miRNAs [31–33] also participate in myofibroblast activation and fibrosis. A review of epigenetic mechanisms regulating renal fibrosis was written by Tampe and Zeisburg in 2012 [89].
Systemic Sclerosis
System sclerosis (SSc) or scleroderma is a rheumatologic disease that causes systemic tissue fibrosis. The current hypothesis for the persistence of activated myofibroblasts in SSc is through profibrotic cytokine stimulation of epigenetic modifications in fibroblasts. In a recent review by Bergmann and Distler in 2017, the authors provide a thorough summary of DNA methylation, histone modifications, microRNAs, and long noncoding RNAs involved in myofibroblast activation along with a list of DNA methyltransferase inhibitors in different phases of drug development [44].
Eye
Epigenetic modifiers play a role in ocular disorders such as pterygia or pterygium, which is a common disease of the eye surface resulting in an abnormal triangular-shaped, fibrotic connective tissue and conjunctival epithelial growth implicating the involvement of fibroblasts [90]. Uncontrolled fibroblast activation and pro-fibrotic signaling causes a pterygium to migrate from the periphery to the center of the cornea causing irritation and can harm vision. Pterygia pathogenesis is multi-factorial and pathways involving fibroblasts and myofibroblasts are not well understood. A few epigenetic mechanisms have been described including aberrant DNA methylation [91] and miRNAs [92]. As surgical treatment is the current intervention of choice, targeting ocular (myo)fibroblasts with an epigenetic therapy is a hopeful avenue to avoid recurrence and potential side effects of eye surgery.
Therapeutic Targets
The successful application of medications that inhibit epigenetic modifications in cancer and the neoplastic-like behavior of myofibroblasts that drives fibrosis has sparked investigations into epigenetic targets that alter myofibroblast behavior. In vitro and a few in vivo studies have established the benefit of pharmacologic therapies that inhibit DNA methylation and histone deacetylation and methylation in the suppression of fibrosis-promoting myofibroblasts [4, 63, 93–101]. Additionally, a small molecule inhibitor of G9a, a lysine methyltransferase, attenuates fibroblast activation [16]; a bromodomain inhibitor prevents fibrosis after intratracheal bleomycin and reduces proliferation and contractile activity in cultured lung fibroblasts [8]; and a BRD4 inhibitor can reverse pre-existing liver fibrosis in a CCl4 mouse model [84].
As fibroblasts and myofibroblasts are the central mediators of fibrosis, epigenetic modifiers that stimulate their apoptosis are a possible therapeutic option, such as HDAC inhibitors which prevent histone modifications responsible for apoptosis resistance [79, 82]. Combinational therapies that incorporate more than one epigenetic mechanism to synergistically reverse or inhibit pro-fibrotic myofibroblast behavior is a promising route. Epigenetic modifiers may also serve as biomarkers of disease severity. For example, expression profiles of miRs in local and diffuse cutaneous SSc [44] and DNA methylation of the PPAR-γ promoter in fibrosis secondary to nonalcoholic fatty liver disease [102].
Future Research
“Epi-drugs” that target active myofibroblasts in fibrosing disorders are a promising direction in the treatment of a myriad of diseases; however, we are far from a comprehensive understanding of how epigenetic modulators influence each other and myofibroblast behavior. This is further complicated by organ and disease-specific variances in epigenetic networks. Future studies must also help us better understand myofibroblast origin and subpopulations.
Regulatory apparatuses involved in disease can be explored by mapping chromatin features to locate tissue-specific enhancers which work with promoters to regulate gene expression [103]. Emerging high throughput single-cell assays that utilize combinatorial indexing, microfluidics, droplet-based methods in combination with new bioinformatics pipelines can better evaluate epigenetic states and trace myofibroblast lineage, possibly identifying rare subpopulations [104]. Reconstructing “epigenetic landscapes” can elucidate candidate transcription factors for reprogramming and modulating cell fate [105]. Recent elegant work in macrophages illustrates how local environments influence enhancer utilization to drive context-dependent heterogeneity of macrophage phenotypes [106]; the same may be true of fibroblasts. Current large-scale efforts to develop multi-“omic” maps of human tissues and organs will provide important knowledge about how normal cell phenotypes are developed and maintained. Another interesting layer to the regulation of gene expression is how local environmental cues (e.g., mechanical stiffness) are transmitted to the nucleus and force-induced changes effect cell signaling and gene transcription [107]. This needs to be further explored as mechanical stress is a key driving factor of myofibroblast activation.
With the advent of bacterial clustered regularly interspaced short palindromic repeats (CRISPRs) system, genome engineering and editing has rapidly progressed and studies pursuing epigenome editing have surfaced including CRISPR-based targeted demethylation [108]. Targeted DNA demethylation is also possible through fusion of a plant 5-methylcytosine DNA glycosylase to sequence-specific DNA binding domains [109]. Applications of targeted epigenome editing would greatly enhance our understanding of how epigenetic modifications regulate gene expression in specific tissues and diseases, translating into targeted tissue and lesion specific anti-myofibroblastic therapies that will have immense clinical benefit.
Overall, the findings presented in this review highlight the complex epigenetic paradigms that directs cell differentiation into pathologic myofibroblast phenotypes in many fibrotic diseases. A more comprehensive understanding of the organ-specific epigenetic mechanisms involved in fibrosis, and of how to target them for therapeutic benefit without significant off-target effects, will facilitate the development of novel antifibrotic therapies.
Acknowledgments
Thu Elizabeth Duong is a Fellow in the Pediatric Scientist Development Program. This project was supported by Award Number K12-HD000850 from the Eunice Kennedy Shriver National Institute of Child Health and Human Development. The work in this publication was supported in part by the National Heart, Lung, and Blood Institute (NHLBI) of the National Institutes of Health (NIH) under Award Number U01HL122626 for J.S.H., as well as 1R01HL111169.
Footnotes
SpringerLink Header: Activated Myofibroblasts and Fibrosis in Various Organs (Tatiana Kisseleva and Youhua Liu, Section Editors)
Compliance with Ethics Guidelines
Conflict of Interest
Dr. Hagood reports personal fees from Kyowa Hakko Kirin, Co., Ltd., outside the submitted work.
Thu Elizabeth Duong declares no conflicts of interest.
Human and Animal Rights and Informed Consent
This article does not contain any studies with human or animal subjects performed by any of the authors
References
Recently published papers of particular interest have been highlighted as:
• Of importance
•• Of major importance
- 1•.El Taghdouini A, van Grunsven LA. Epigenetic regulation of hepatic stellate cell activation and liver fibrosis. Expert review of gastroenterology & hepatology. 2016;10(12):1397–408. doi: 10.1080/17474124.2016.1251309. Review of epigenetic regulation of HSC activation in liver fibrosis. [DOI] [PubMed] [Google Scholar]
- 2.Shiga K, Hara M, Nagasaki T, Sato T, Takahashi H, Takeyama H. Cancer-Associated Fibroblasts: Their Characteristics and Their Roles in Tumor Growth. Cancers (Basel) 2015;7(4):2443–58. doi: 10.3390/cancers7040902. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3•.Hinz B. Myofibroblasts. Exp Eye Res. 2016;142:56–70. doi: 10.1016/j.exer.2015.07.009. Comprehensive review of current state of myofibroblast literature with an emphasis on myofibroblast involvement in ocular fibrosis. [DOI] [PubMed] [Google Scholar]
- 4.Mann J, Chu DC, Maxwell A, Oakley F, Zhu NL, Tsukamoto H, et al. MeCP2 controls an epigenetic pathway that promotes myofibroblast transdifferentiation and fibrosis. Gastroenterology. 2010;138(2):705–14. 14.e1–4. doi: 10.1053/j.gastro.2009.10.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Kohli RM, Zhang Y. TET enzymes, TDG and the dynamics of DNA demethylation. Nature. 2013;502(7472):472–9. doi: 10.1038/nature12750. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Jang HS, Shin WJ, Lee JE, Do JT. CpG and Non-CpG Methylation in Epigenetic Gene Regulation and Brain Function. Genes. 2017;8(6) doi: 10.3390/genes8060148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Ghosh AK, Bhattacharyya S, Lafyatis R, Farina G, Yu J, Thimmapaya B, et al. p300 is elevated in systemic sclerosis and its expression is positively regulated by TGF-beta: epigenetic feed-forward amplification of fibrosis. The Journal of investigative dermatology. 2013;133(5):1302–10. doi: 10.1038/jid.2012.479. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Tang X, Peng R, Ren Y, Apparsundaram S, Deguzman J, Bauer CM, et al. BET bromodomain proteins mediate downstream signaling events following growth factor stimulation in human lung fibroblasts and are involved in bleomycin-induced pulmonary fibrosis. Molecular pharmacology. 2013;83(1):283–93. doi: 10.1124/mol.112.081661. [DOI] [PubMed] [Google Scholar]
- 9.Glenisson W, Castronovo V, Waltregny D. Histone deacetylase 4 is required for TGFbeta1-induced myofibroblastic differentiation. Biochimica et biophysica acta. 2007;1773(10):1572–82. doi: 10.1016/j.bbamcr.2007.05.016. [DOI] [PubMed] [Google Scholar]
- 10.Coward WR, Watts K, Feghali-Bostwick CA, Knox A, Pang L. Defective Histone Acetylation Is Responsible for the Diminished Expression of Cyclooxygenase 2 in Idiopathic Pulmonary Fibrosis. Molecular and Cellular Biology. 2009;29(15):4325–39. doi: 10.1128/mcb.01776-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Wang Y, Fan PS, Kahaleh B. Association between enhanced type I collagen expression and epigenetic repression of the FLI1 gene in scleroderma fibroblasts. Arthritis and rheumatism. 2006;54(7):2271–9. doi: 10.1002/art.21948. [DOI] [PubMed] [Google Scholar]
- 12.Noda S, Asano Y, Nishimura S, Taniguchi T, Fujiu K, Manabe I, et al. Simultaneous downregulation of KLF5 and Fli1 is a key feature underlying systemic sclerosis. Nature communications. 2014;5:5797. doi: 10.1038/ncomms6797. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Coward WR, Watts K, Feghali-Bostwick CA, Jenkins G, Pang L. Repression of IP-10 by interactions between histone deacetylation and hypermethylation in idiopathic pulmonary fibrosis. Molecular and Cellular Biology. 2010;30(12):2874–86. doi: 10.1128/mcb.01527-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Jiang Y, Wang S, Zhao Y, Lin C, Zhong F, Jin L, et al. Histone H3K9 demethylase JMJD1A modulates hepatic stellate cells activation and liver fibrosis by epigenetically regulating peroxisome proliferator-activated receptor gamma. FASEB journal : official publication of the Federation of American Societies for Experimental Biology. 2015;29(5):1830–41. doi: 10.1096/fj.14-251751. [DOI] [PubMed] [Google Scholar]
- 15.Bergmann C, Brandt A, Dees C, Zhang Y, Lin NY, Chen C, et al. Jumonji Domain Containing Protein 3 (JMJD3) As a Novel Epigenetic Mechanism of Fibroblast Activation By Regulation of Fra-2 [abstract]. American College of Rhuematology Annual Meeting; November 14, 2016; 2016. [Google Scholar]
- 16.Coward WR, Feghali-Bostwick CA, Jenkins G, Knox AJ, Pang L. A central role for G9a and EZH2 in the epigenetic silencing of cyclooxygenase-2 in idiopathic pulmonary fibrosis. FASEB journal : official publication of the Federation of American Societies for Experimental Biology. 2014;28(7):3183–96. doi: 10.1096/fj.13-241760. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Beermann J, Piccoli MT, Viereck J, Thum T. Non-coding RNAs in Development and Disease: Background, Mechanisms, and Therapeutic Approaches. Physiological reviews. 2016;96(4):1297–325. doi: 10.1152/physrev.00041.2015. [DOI] [PubMed] [Google Scholar]
- 18.Song R, Fullerton DA, Ao L, Zhao KS, Reece TB, Cleveland JC, Jr, et al. Altered MicroRNA Expression Is Responsible for the Pro-Osteogenic Phenotype of Interstitial Cells in Calcified Human Aortic Valves. Journal of the American Heart Association. 2017;6(4) doi: 10.1161/jaha.116.005364. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.van Rooij E, Sutherland LB, Thatcher JE, DiMaio JM, Naseem RH, Marshall WS, et al. Dysregulation of microRNAs after myocardial infarction reveals a role of miR-29 in cardiac fibrosis. Proceedings of the National Academy of Sciences of the United States of America. 2008;105(35):13027–32. doi: 10.1073/pnas.0805038105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.da Costa Martins PA, Salic K, Gladka MM, Armand AS, Leptidis S, el Azzouzi H, et al. MicroRNA-199b targets the nuclear kinase Dyrk1a in an auto-amplification loop promoting calcineurin/NFAT signalling. Nature cell biology. 2010;12(12):1220–7. doi: 10.1038/ncb2126. [DOI] [PubMed] [Google Scholar]
- 21.Roderburg C, Urban GW, Bettermann K, Vucur M, Zimmermann H, Schmidt S, et al. Micro-RNA profiling reveals a role for miR-29 in human and murine liver fibrosis. Hepatology (Baltimore, Md) 2011;53(1):209–18. doi: 10.1002/hep.23922. [DOI] [PubMed] [Google Scholar]
- 22.Roderburg C, Luedde M, Vargas Cardenas D, Vucur M, Mollnow T, Zimmermann HW, et al. miR-133a mediates TGF-beta-dependent derepression of collagen synthesis in hepatic stellate cells during liver fibrosis. Journal of hepatology. 2013;58(4):736–42. doi: 10.1016/j.jhep.2012.11.022. [DOI] [PubMed] [Google Scholar]
- 23.Li ZJ, Ou-Yang PH, Han XP. Profibrotic effect of miR-33a with Akt activation in hepatic stellate cells. Cellular signalling. 2014;26(1):141–8. doi: 10.1016/j.cellsig.2013.09.018. [DOI] [PubMed] [Google Scholar]
- 24.Yu F, Guo Y, Chen B, Dong P, Zheng J. MicroRNA-17-5p activates hepatic stellate cells through targeting of Smad7. Lab Invest. 2015;95(7):781–9. doi: 10.1038/labinvest.2015.58. [DOI] [PubMed] [Google Scholar]
- 25.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):1589–97. doi: 10.1084/jem.20100035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.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):220–9. doi: 10.1164/rccm.200911-1698OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.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):287–94. doi: 10.1165/rcmb.2010-0323OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Xiao J, Meng XM, Huang XR, Chung AC, Feng YL, Hui DS, et al. miR-29 inhibits bleomycin-induced pulmonary fibrosis in mice. Mol Ther. 2012;20(6):1251–60. doi: 10.1038/mt.2012.36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Dakhlallah D, Batte K, Wang Y, Cantemir-Stone CZ, Yan P, Nuovo G, et al. Epigenetic regulation of miR-17~92 contributes to the pathogenesis of pulmonary fibrosis. American journal of respiratory and critical care medicine. 2013;187(4):397–405. doi: 10.1164/rccm.201205-0888OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Lino Cardenas CL, Henaoui IS, Courcot E, Roderburg C, Cauffiez C, Aubert S, et al. miR-199a-5p Is upregulated during fibrogenic response to tissue injury and mediates TGFbeta-induced lung fibroblast activation by targeting caveolin-1. PLoS genetics. 2013;9(2):e1003291. doi: 10.1371/journal.pgen.1003291. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Krupa A, Jenkins R, Luo DD, Lewis A, Phillips A, Fraser D. Loss of MicroRNA-192 promotes fibrogenesis in diabetic nephropathy. Journal of the American Society of Nephrology : JASN. 2010;21(3):438–47. doi: 10.1681/asn.2009050530. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Qin W, Chung AC, Huang XR, Meng XM, Hui DS, Yu CM, et al. TGF-beta/Smad3 signaling promotes renal fibrosis by inhibiting miR-29. Journal of the American Society of Nephrology : JASN. 2011;22(8):1462–74. doi: 10.1681/asn.2010121308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Ramdas V, McBride M, Denby L, Baker AH. Canonical transforming growth factor-beta signaling regulates disintegrin metalloprotease expression in experimental renal fibrosis via miR-29. The American journal of pathology. 2013;183(6):1885–96. doi: 10.1016/j.ajpath.2013.08.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Kajihara I, Jinnin M, Yamane K, Makino T, Honda N, Igata T, et al. Increased accumulation of extracellular thrombospondin-2 due to low degradation activity stimulates type I collagen expression in scleroderma fibroblasts. The American journal of pathology. 2012;180(2):703–14. doi: 10.1016/j.ajpath.2011.10.030. [DOI] [PubMed] [Google Scholar]
- 35.Ciechomska M, O’Reilly S, Suwara M, Bogunia-Kubik K, van Laar JM. MiR-29a reduces TIMP-1 production by dermal fibroblasts via targeting TGF-beta activated kinase 1 binding protein 1, implications for systemic sclerosis. PloS one. 2014;9(12):e115596. doi: 10.1371/journal.pone.0115596. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Maurer B, Stanczyk J, Jungel A, Akhmetshina A, Trenkmann M, Brock M, et al. MicroRNA-29, a key regulator of collagen expression in systemic sclerosis. Arthritis and rheumatism. 2010;62(6):1733–43. doi: 10.1002/art.27443. [DOI] [PubMed] [Google Scholar]
- 37.Zhu H, Li Y, Qu S, Luo H, Zhou Y, Wang Y, et al. MicroRNA expression abnormalities in limited cutaneous scleroderma and diffuse cutaneous scleroderma. Journal of clinical immunology. 2012;32(3):514–22. doi: 10.1007/s10875-011-9647-y. [DOI] [PubMed] [Google Scholar]
- 38.Nakashima T, Jinnin M, Yamane K, Honda N, Kajihara I, Makino T, et al. Impaired IL-17 signaling pathway contributes to the increased collagen expression in scleroderma fibroblasts. Journal of immunology (Baltimore, Md : 1950) 2012;188(8):3573–83. doi: 10.4049/jimmunol.1100591. [DOI] [PubMed] [Google Scholar]
- 39.Honda N, Jinnin M, Kira-Etoh T, Makino K, Kajihara I, Makino T, et al. miR-150 down-regulation contributes to the constitutive type I collagen overexpression in scleroderma dermal fibroblasts via the induction of integrin beta3. The American journal of pathology. 2013;182(1):206–16. doi: 10.1016/j.ajpath.2012.09.023. [DOI] [PubMed] [Google Scholar]
- 40.Honda N, Jinnin M, Kajihara I, Makino T, Makino K, Masuguchi S, et al. TGF-beta-mediated downregulation of microRNA-196a contributes to the constitutive upregulated type I collagen expression in scleroderma dermal fibroblasts. Journal of immunology (Baltimore, Md : 1950) 2012;188(7):3323–31. doi: 10.4049/jimmunol.1100876. [DOI] [PubMed] [Google Scholar]
- 41.Li H, Yang R, Fan X, Gu T, Zhao Z, Chang D, et al. MicroRNA array analysis of microRNAs related to systemic scleroderma. Rheumatol Int. 2012;32(2):307–13. doi: 10.1007/s00296-010-1615-y. [DOI] [PubMed] [Google Scholar]
- 42.Yan Q, Chen J, Li W, Bao C, Fu Q. Targeting miR-155 to Treat Experimental Scleroderma. Scientific reports. 2016;6:20314. doi: 10.1038/srep20314. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Christmann RB, Wooten A, Sampaio-Barros P, Borges CL, Carvalho CR, Kairalla RA, et al. miR-155 in the progression of lung fibrosis in systemic sclerosis. Arthritis Res Ther. 2016;18(1):155. doi: 10.1186/s13075-016-1054-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44•.Bergmann C, Distler JH. Epigenetic factors as drivers of fibrosis in systemic sclerosis. Epigenomics. 2017;9(4):463–77. doi: 10.2217/epi-2016-0150. Review of epigenetic factors involved in systemic sclerosis. [DOI] [PubMed] [Google Scholar]
- 45.O’Reilly S, Ciechomska M, Fullard N, Przyborski S, van Laar JM. IL-13 mediates collagen deposition via STAT6 and microRNA-135b: a role for epigenetics. Scientific reports. 2016;6:25066. doi: 10.1038/srep25066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Xu X, Tan X, Tampe B, Nyamsuren G, Liu X, Maier LS, et al. Epigenetic balance of aberrant Rasal1 promoter methylation and hydroxymethylation regulates cardiac fibrosis. Cardiovascular research. 2015;105(3):279–91. doi: 10.1093/cvr/cvv015. [DOI] [PubMed] [Google Scholar]
- 47.Xu X, Tan X, Hulshoff MS, Wilhelmi T, Zeisberg M, Zeisberg EM. Hypoxia-induced endothelial-mesenchymal transition is associated with RASAL1 promoter hypermethylation in human coronary endothelial cells. FEBS letters. 2016;590(8):1222–33. doi: 10.1002/1873-3468.12158. [DOI] [PubMed] [Google Scholar]
- 48.Watson CJ, Collier P, Tea I, Neary R, Watson JA, Robinson C, et al. Hypoxia-induced epigenetic modifications are associated with cardiac tissue fibrosis and the development of a myofibroblast-like phenotype. Hum Mol Genet. 2014;23(8):2176–88. doi: 10.1093/hmg/ddt614. [DOI] [PubMed] [Google Scholar]
- 49.Tao H, Yang JJ, Shi KH, Deng ZY, Li J. DNA methylation in cardiac fibrosis: new advances and perspectives. Toxicology. 2014;323:125–9. doi: 10.1016/j.tox.2014.07.002. [DOI] [PubMed] [Google Scholar]
- 50.Tao H, Yang JJ, Hu W, Shi KH, Li J. HDAC6 Promotes Cardiac Fibrosis Progression through Suppressing RASSF1A Expression. Cardiology. 2016;133(1):18–26. doi: 10.1159/000438781. [DOI] [PubMed] [Google Scholar]
- 51.Williams SM, Golden-Mason L, Ferguson BS, Douglas KB, Cavasin MA, Demos-Davies K, et al. Class I HDACs regulate angiotensin II-dependent cardiac fibrosis via fibroblasts and circulating fibrocytes. J Mol Cell Cardiol. 2013;67 doi: 10.1016/j.yjmcc.2013.12.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Nural-Guvener H, Zakharova L, Feehery L, Sljukic S, Gaballa M. Anti-Fibrotic Effects of Class I HDAC Inhibitor, Mocetinostat Is Associated with IL-6/Stat3 Signaling in Ischemic Heart Failure. International journal of molecular sciences. 2015;16(5):11482–99. doi: 10.3390/ijms160511482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Nural-Guvener HF, Zakharova L, Nimlos J, Popovic S, Mastroeni D, Gaballa MA. HDAC class I inhibitor. Fibrogenesis & Tissue Repair. 2014;7(1):10. doi: 10.1186/1755-1536-7-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Kao YH, Liou JP, Chung CC, Lien GS, Kuo CC, Chen SA, et al. Histone deacetylase inhibition improved cardiac functions with direct antifibrotic activity in heart failure. Int J Cardiol. 2013;168 doi: 10.1016/j.ijcard.2013.07.111. [DOI] [PubMed] [Google Scholar]
- 55.Liu F, Levin MD, Petrenko NB, Lu MM, Wang T, Yuan LJ, et al. Histone-deacetylase inhibition reverses atrial arrhythmia inducibility and fibrosis in cardiac hypertrophy independent of angiotensin. J Mol Cell Cardiol. 2008;45 doi: 10.1016/j.yjmcc.2008.08.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56•.Grimaldi V, De Pascale MR, Zullo A, Soricelli A, Infante T, Mancini FP, et al. Evidence of epigenetic tags in cardiac fibrosis. Journal of cardiology. 2017;69(2):401–8. doi: 10.1016/j.jjcc.2016.10.004. A review of epigenetic changes involved in cardiac fibrosis. [DOI] [PubMed] [Google Scholar]
- 57.Stratton MS, McKinsey TA. Epigenetic regulation of cardiac fibrosis. J Mol Cell Cardiol. 2016;92:206–13. doi: 10.1016/j.yjmcc.2016.02.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Olsen MB, Hildrestrand GA, Scheffler K, Vinge LE, Alfsnes K, Palibrk V, et al. NEIL3-Dependent Regulation of Cardiac Fibroblast Proliferation Prevents Myocardial Rupture. Cell reports. 2017;18(1):82–92. doi: 10.1016/j.celrep.2016.12.009. [DOI] [PubMed] [Google Scholar]
- 59.Liu AC, Joag VR, Gotlieb AI. The emerging role of valve interstitial cell phenotypes in regulating heart valve pathobiology. The American journal of pathology. 2007;171(5):1407–18. doi: 10.2353/ajpath.2007.070251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Renaud L, Harris LG, Mani SK, Kasiganesan H, Chou JC, Baicu CF, et al. HDACs Regulate miR-133a Expression in Pressure Overload-Induced Cardiac Fibrosis. Circulation Heart failure. 2015;8(6):1094–104. doi: 10.1161/circheartfailure.114.001781. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Karin D, Koyama Y, Brenner D, Kisseleva T. The characteristics of activated portal fibroblasts/myofibroblasts in liver fibrosis. Differentiation; research in biological diversity. 2016;92(3):84–92. doi: 10.1016/j.diff.2016.07.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Nwosu ZC, Alborzinia H, Wolfl S, Dooley S, Liu Y. Evolving Insights on Metabolism, Autophagy, and Epigenetics in Liver Myofibroblasts. Frontiers in physiology. 2016;7:191. doi: 10.3389/fphys.2016.00191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Mann J, Oakley F, Akiboye F, Elsharkawy A, Thorne AW, Mann DA. Regulation of myofibroblast transdifferentiation by DNA methylation and MeCP2: implications for wound healing and fibrogenesis. Cell death and differentiation. 2007;14(2):275–85. doi: 10.1038/sj.cdd.4401979. [DOI] [PubMed] [Google Scholar]
- 64.Wu Y, Bu F, Yu H, Li W, Huang C, Meng X, et al. Methylation of Septin9 mediated by DNMT3a enhances hepatic stellate cells activation and liver fibrogenesis. Toxicology and applied pharmacology. 2017;315:35–49. doi: 10.1016/j.taap.2016.12.002. [DOI] [PubMed] [Google Scholar]
- 65.Shi C, Li G, Tong Y, Deng Y, Fan J. Role of CTGF gene promoter methylation in the development of hepatic fibrosis. American journal of translational research. 2016;8(1):125–32. [PMC free article] [PubMed] [Google Scholar]
- 66.Bian EB, Huang C, Wang H, Chen XX, Zhang L, Lv XW, et al. Repression of Smad7 mediated by DNMT1 determines hepatic stellate cell activation and liver fibrosis in rats. Toxicology letters. 2014;224(2):175–85. doi: 10.1016/j.toxlet.2013.10.038. [DOI] [PubMed] [Google Scholar]
- 67.Bian E-B, Huang C, Ma T-T, Tao H, Zhang H, Cheng C, et al. DNMT1-mediated PTEN hypermethylation confers hepatic stellate cell activation and liver fibrogenesis in rats. Toxicology and applied pharmacology. 2012;264(1):13–22. doi: 10.1016/j.taap.2012.06.022. [DOI] [PubMed] [Google Scholar]
- 68.Yang JJ, Tao H, Huang C, Shi KH, Ma TT, Bian EB, et al. DNA methylation and MeCP2 regulation of PTCH1 expression during rats hepatic fibrosis. Cellular signalling. 2013;25(5):1202–11. doi: 10.1016/j.cellsig.2013.01.005. [DOI] [PubMed] [Google Scholar]
- 69.Qin L, Han YP. Epigenetic repression of matrix metalloproteinases in myofibroblastic hepatic stellate cells through histone deacetylases 4: implication in tissue fibrosis. The American journal of pathology. 2010;177(4):1915–28. doi: 10.2353/ajpath.2010.100011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Pannem RR, Dorn C, Hellerbrand C, Massoumi R. Cylindromatosis gene CYLD regulates hepatocyte growth factor expression in hepatic stellate cells through interaction with histone deacetylase 7. Hepatology (Baltimore, Md) 2014;60(3):1066–81. doi: 10.1002/hep.27209. [DOI] [PubMed] [Google Scholar]
- 71.Page A, Paoli PP, Hill SJ, Howarth R, Wu R, Kweon SM, et al. Alcohol directly stimulates epigenetic modifications in hepatic stellate cells. Journal of hepatology. 2015;62(2):388–97. doi: 10.1016/j.jhep.2014.09.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Ding N, Yu RT, Subramaniam N, Sherman MH, Wilson C, Rao R, et al. A vitamin D receptor/SMAD genomic circuit gates hepatic fibrotic response. Cell. 2013;153(3):601–13. doi: 10.1016/j.cell.2013.03.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Wang Y, Chen C, Deng Z, Bian E, Huang C, Lei T, et al. Repression of TSC1/TSC2 mediated by MeCP2 regulates human embryo lung fibroblast cell differentiation and proliferation. International journal of biological macromolecules. 2017;96:578–88. doi: 10.1016/j.ijbiomac.2016.12.062. [DOI] [PubMed] [Google Scholar]
- 74•.Tzouvelekis A, Kaminski N. Epigenetics in idiopathic pulmonary fibrosis. Biochemistry and cell biology = Biochimie et biologie cellulaire. 2015;93(2):159–70. doi: 10.1139/bcb-2014-0126. Review of epigenetic mechanisms involved in IPF. [DOI] [PubMed] [Google Scholar]
- 75.Pandit KV, Milosevic J, Kaminski N. MicroRNAs in idiopathic pulmonary fibrosis. Translational research : the journal of laboratory and clinical medicine. 2011;157(4):191–9. doi: 10.1016/j.trsl.2011.01.012. [DOI] [PubMed] [Google Scholar]
- 76.Sanders YY, Pardo A, Selman M, Nuovo GJ, Tollefsbol TO, Siegal GP, et al. Thy-1 Promoter Hypermethylation: A Novel Epigenetic Pathogenic Mechanism in Pulmonary Fibrosis. American journal of respiratory cell and molecular biology. 2008;39(5):610–8. doi: 10.1165/rcmb.2007-0322OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Sanders YY, Ambalavanan N, Halloran B, Zhang X, Liu H, Crossman DK, et al. Altered DNA methylation profile in idiopathic pulmonary fibrosis. American journal of respiratory and critical care medicine. 2012;186(6):525–35. doi: 10.1164/rccm.201201-0077OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Sanders YY, Tollefsbol TO, Varisco BM, Hagood JS. Epigenetic regulation of thy-1 by histone deacetylase inhibitor in rat lung fibroblasts. American journal of respiratory cell and molecular biology. 2011;45(1):16–23. doi: 10.1165/rcmb.2010-0154OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Huang SK, Scruggs AM, Donaghy J, Horowitz JC, Zaslona Z, Przybranowski S, et al. Histone modifications are responsible for decreased Fas expression and apoptosis resistance in fibrotic lung fibroblasts. Cell death & disease. 2013;4:e621. doi: 10.1038/cddis.2013.146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Korfei M, Skwarna S, Henneke I, MacKenzie B, Klymenko O, Saito S, et al. Aberrant expression and activity of histone deacetylases in sporadic idiopathic pulmonary fibrosis. Thorax. 2015;70(11):1022–32. doi: 10.1136/thoraxjnl-2014-206411. [DOI] [PubMed] [Google Scholar]
- 81.Guo W, Shan B, Klingsberg RC, Qin X, Lasky JA. Abrogation of TGF-beta1-induced fibroblast-myofibroblast differentiation by histone deacetylase inhibition. American journal of physiology Lung cellular and molecular physiology. 2009;297(5):L864–70. doi: 10.1152/ajplung.00128.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Sanders YY, Hagood JS, Liu H, Zhang W, Ambalavanan N, Thannickal VJ. Histone deacetylase inhibition promotes fibroblast apoptosis and ameliorates pulmonary fibrosis in mice. The European respiratory journal. 2014;43(5):1448–58. doi: 10.1183/09031936.00095113. [DOI] [PubMed] [Google Scholar]
- 83.Davies ER, Haitchi HM, Thatcher TH, Sime PJ, Kottmann RM, Ganesan A, et al. Spiruchostatin A inhibits proliferation and differentiation of fibroblasts from patients with pulmonary fibrosis. American journal of respiratory cell and molecular biology. 2012;46(5):687–94. doi: 10.1165/rcmb.2011-0040OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Tang X, Peng R, Phillips JE, Deguzman J, Ren Y, Apparsundaram S, et al. Assessment of Brd4 inhibition in idiopathic pulmonary fibrosis lung fibroblasts and in vivo models of lung fibrosis. The American journal of pathology. 2013;183(2):470–9. doi: 10.1016/j.ajpath.2013.04.020. [DOI] [PubMed] [Google Scholar]
- 85.Bechtel W, McGoohan S, Zeisberg EM, Muller GA, Kalbacher H, Salant DJ, et al. Methylation determines fibroblast activation and fibrogenesis in the kidney. Nature medicine. 2010;16(5):544–50. doi: 10.1038/nm.2135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Chang YT, Yang CC, Pan SY, Chou YH, Chang FC, Lai CF, et al. DNA methyltransferase inhibition restores erythropoietin production in fibrotic murine kidneys. The Journal of clinical investigation. 2016;126(2):721–31. doi: 10.1172/jci82819. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Pang M, Kothapally J, Mao H, Tolbert E, Ponnusamy M, Chin YE, et al. Inhibition of histone deacetylase activity attenuates renal fibroblast activation and interstitial fibrosis in obstructive nephropathy. American journal of physiology Renal physiology. 2009;297(4):F996–f1005. doi: 10.1152/ajprenal.00282.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Ponnusamy M, Zhou X, Yan Y, Tang J, Tolbert E, Zhao TC, et al. Blocking sirtuin 1 and 2 inhibits renal interstitial fibroblast activation and attenuates renal interstitial fibrosis in obstructive nephropathy. The Journal of pharmacology and experimental therapeutics. 2014;350(2):243–56. doi: 10.1124/jpet.113.212076. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Tampe D, Zeisberg M. A primer on the epigenetics of kidney fibrosis. Minerva medica. 2012;103(4):267–78. [PubMed] [Google Scholar]
- 90.Kim KW, Park SH, Kim JC. Fibroblast biology in pterygia. Exp Eye Res. 2016;142:32–9. doi: 10.1016/j.exer.2015.01.010. [DOI] [PubMed] [Google Scholar]
- 91.Riau AK, Wong TT, Lan W, Finger SN, Chaurasia SS, Hou AH, et al. Aberrant DNA methylation of matrix remodeling and cell adhesion related genes in pterygium. PloS one. 2011;6(2):e14687. doi: 10.1371/journal.pone.0014687. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Engelsvold DH, Utheim TP, Olstad OK, Gonzalez P, Eidet JR, Lyberg T, et al. miRNA and mRNA expression profiling identifies members of the miR-200 family as potential regulators of epithelial-mesenchymal transition in pterygium. Exp Eye Res. 2013;115:189–98. doi: 10.1016/j.exer.2013.07.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Page A, Paoli P, Moran Salvador E, White S, French J, Mann J. Hepatic stellate cell transdifferentiation involves genome-wide remodeling of the DNA methylation landscape. Journal of hepatology. 2016;64(3):661–73. doi: 10.1016/j.jhep.2015.11.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Niki T, Rombouts K, De Bleser P, De Smet K, Rogiers V, Schuppan D, et al. A histone deacetylase inhibitor, trichostatin A, suppresses myofibroblastic differentiation of rat hepatic stellate cells in primary culture. Hepatology (Baltimore, Md) 1999;29(3):858–67. doi: 10.1002/hep.510290328. [DOI] [PubMed] [Google Scholar]
- 95.Park KC, Park JH, Jeon JY, Kim SY, Kim JM, Lim CY, et al. A new histone deacetylase inhibitor improves liver fibrosis in BDL rats through suppression of hepatic stellate cells. British Journal of Pharmacology. 2014;171(21):4820–30. doi: 10.1111/bph.12590. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Liu Y, Wang Z, Wang J, Lam W, Kwong S, Li F, et al. A histone deacetylase inhibitor, largazole, decreases liver fibrosis and angiogenesis by inhibiting transforming growth factor-β and vascular endothelial growth factor signalling. Liver International. 2013;33(4):504–15. doi: 10.1111/liv.12034. [DOI] [PubMed] [Google Scholar]
- 97.Mannaerts I, Nuytten NR, Rogiers V, Vanderkerken K, van Grunsven LA, Geerts A. Chronic administration of valproic acid inhibits activation of mouse hepatic stellate cells in vitro and in vivo. Hepatology (Baltimore, Md) 2010;51(2):603–14. doi: 10.1002/hep.23334. [DOI] [PubMed] [Google Scholar]
- 98.Zeybel M, Luli S, Sabater L, Hardy T, Oakley F, Leslie J, et al. A Proof-of-Concept for Epigenetic Therapy of Tissue Fibrosis: Inhibition of Liver Fibrosis Progression by 3-Deazaneplanocin A. Mol Ther. 2017;25(1):218–31. doi: 10.1016/j.ymthe.2016.10.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Kim YS, Kang WS, Kwon JS, Hong MH, Jeong HY, Jeong HC, et al. Protective role of 5-azacytidine on myocardial infarction is associated with modulation of macrophage phenotype and inhibition of fibrosis. Journal of cellular and molecular medicine. 2014;18(6):1018–27. doi: 10.1111/jcmm.12248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Watson CJ, Horgan S, Neary R, Glezeva N, Tea I, Corrigan N, et al. Epigenetic Therapy for the Treatment of Hypertension-Induced Cardiac Hypertrophy and Fibrosis. Journal of Cardiovascular Pharmacology and Therapeutics. 2016;21(1):127–37. doi: 10.1177/1074248415591698. [DOI] [PubMed] [Google Scholar]
- 101.Kaimori A, Potter JJ, Choti M, Ding Z, Mezey E, Koteish AA. Histone deacetylase inhibition suppresses the transforming growth factor beta1-induced epithelial-to-mesenchymal transition in hepatocytes. Hepatology (Baltimore, Md) 2010;52(3):1033–45. doi: 10.1002/hep.23765. [DOI] [PubMed] [Google Scholar]
- 102.Hardy T, Zeybel M, Day CP, Dipper C, Masson S, McPherson S, et al. Plasma DNA methylation: a potential biomarker for stratification of liver fibrosis in non-alcoholic fatty liver disease. Gut. 2017;66(7):1321–8. doi: 10.1136/gutjnl-2016-311526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Bowman SK. Discovering enhancers by mapping chromatin features in primary tissue. Genomics. 2015;106(3):140–4. doi: 10.1016/j.ygeno.2015.06.006. [DOI] [PubMed] [Google Scholar]
- 104.Clark SJ, Lee HJ, Smallwood SA, Kelsey G, Reik W. Single-cell epigenomics: powerful new methods for understanding gene regulation and cell identity. Genome Biology. 2016;17(1):72. doi: 10.1186/s13059-016-0944-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Lang AH, Li H, Collins JJ, Mehta P. Epigenetic landscapes explain partially reprogrammed cells and identify key reprogramming genes. PLoS computational biology. 2014;10(8):e1003734. doi: 10.1371/journal.pcbi.1003734. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Glass CK, Natoli G. Molecular control of activation and priming in macrophages. Nature immunology. 2016;17(1):26–33. doi: 10.1038/ni.3306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Dahl KN, Ribeiro AJ, Lammerding J. Nuclear shape, mechanics, and mechanotransduction. Circulation research. 2008;102(11):1307–18. doi: 10.1161/circresaha.108.173989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Xu X, Tao Y, Gao X, Zhang L, Li X, Zou W, et al. A CRISPR-based approach for targeted DNA demethylation. Cell discovery. 2016;2:16009. doi: 10.1038/celldisc.2016.9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Parrilla-Doblas JT, Ariza RR, Roldan-Arjona T. Targeted DNA demethylation in human cells by fusion of a plant 5-methylcytosine DNA glycosylase to a sequence-specific DNA binding domain. Epigenetics. 2017;12(4):296–303. doi: 10.1080/15592294.2017.1294306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Tao H, Yang JJ, Chen ZW, Xu SS, Zhou X, Zhan HY, et al. DNMT3A silencing RASSF1A promotes cardiac fibrosis through upregulation of ERK1/2. Toxicology. 2014;323:42–50. doi: 10.1016/j.tox.2014.06.006. [DOI] [PubMed] [Google Scholar]
- 111.Thum T, Gross C, Fiedler J, Fischer T, Kissler S, Bussen M, et al. MicroRNA-21 contributes to myocardial disease by stimulating MAP kinase signalling in fibroblasts. Nature. 2008;456(7224):980–4. doi: 10.1038/nature07511. [DOI] [PubMed] [Google Scholar]
- 112.Wang YS, Li SH, Guo J, Mihic A, Wu J, Sun L, et al. Role of miR-145 in cardiac myofibroblast differentiation. J Mol Cell Cardiol. 2014;66:94–105. doi: 10.1016/j.yjmcc.2013.08.007. [DOI] [PubMed] [Google Scholar]
- 113.Perugorria MJ, Wilson CL, Zeybel M, Walsh M, Amin S, Robinson S, et al. Histone methyltransferase ASH1 orchestrates fibrogenic gene transcription during myofibroblast transdifferentiation. Hepatology (Baltimore, Md) 2012;56(3):1129–39. doi: 10.1002/hep.25754. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Ding N, Hah N, Yu RT, Sherman MH, Benner C, Leblanc M, et al. BRD4 is a novel therapeutic target for liver fibrosis. Proceedings of the National Academy of Sciences of the United States of America. 2015;112(51):15713–8. doi: 10.1073/pnas.1522163112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Xu H, Wu X, Qin H, Tian W, Chen J, Sun L, et al. Myocardin-Related Transcription Factor A Epigenetically Regulates Renal Fibrosis in Diabetic Nephropathy. Journal of the American Society of Nephrology : JASN. 2015;26(7):1648–60. doi: 10.1681/asn.2014070678. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Dees C, Schlottmann I, Funke R, Distler A, Palumbo-Zerr K, Zerr P, et al. The Wnt antagonists DKK1 and SFRP1 are downregulated by promoter hypermethylation in systemic sclerosis. Annals of the rheumatic diseases. 2014;73(6):1232–9. doi: 10.1136/annrheumdis-2012-203194. [DOI] [PubMed] [Google Scholar]
- 117.Svegliati S, Marrone G, Pezone A, Spadoni T, Grieco A, Moroncini G, et al. Oxidative DNA damage induces the ATM-mediated transcriptional suppression of the Wnt inhibitor WIF-1 in systemic sclerosis and fibrosis. Science signaling. 2014;7(341):ra84. doi: 10.1126/scisignal.2004592. [DOI] [PubMed] [Google Scholar]
- 118.Huber LC, Distler JH, Moritz F, Hemmatazad H, Hauser T, Michel BA, et al. Trichostatin A prevents the accumulation of extracellular matrix in a mouse model of bleomycin-induced skin fibrosis. Arthritis and rheumatism. 2007;56(8):2755–64. doi: 10.1002/art.22759. [DOI] [PubMed] [Google Scholar]
- 119.Hemmatazad H, Rodrigues HM, Maurer B, Brentano F, Pileckyte M, Distler JH, et al. Histone deacetylase 7, a potential target for the antifibrotic treatment of systemic sclerosis. Arthritis and rheumatism. 2009;60(5):1519–29. doi: 10.1002/art.24494. [DOI] [PubMed] [Google Scholar]
- 120.Bhattacharyya S, Ghosh AK, Pannu J, Mori Y, Takagawa S, Chen G, et al. Fibroblast expression of the coactivator p300 governs the intensity of profibrotic response to transforming growth factor beta. Arthritis and rheumatism. 2005;52(4):1248–58. doi: 10.1002/art.20996. [DOI] [PubMed] [Google Scholar]
- 121.Zerr P, Palumbo-Zerr K, Huang J, Tomcik M, Sumova B, Distler O, et al. Sirt1 regulates canonical TGF-beta signalling to control fibroblast activation and tissue fibrosis. Annals of the rheumatic diseases. 2016;75(1):226–33. doi: 10.1136/annrheumdis-2014-205740. [DOI] [PubMed] [Google Scholar]
- 122.Wei J, Ghosh AK, Chu H, Fang F, Hinchcliff ME, Wang J, et al. The Histone Deacetylase Sirtuin 1 Is Reduced in Systemic Sclerosis and Abrogates Fibrotic Responses by Targeting Transforming Growth Factor beta Signaling. Arthritis & rheumatology (Hoboken, NJ) 2015;67(5):1323–34. doi: 10.1002/art.39061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Akamata K, Wei J, Bhattacharyya M, Cheresh P, Bonner MY, Arbiser JL, et al. SIRT3 is attenuated in systemic sclerosis skin and lungs, and its pharmacologic activation mitigates organ fibrosis. Oncotarget. 2016;7(43):69321–36. doi: 10.18632/oncotarget.12504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Kramer M, Dees C, Huang J, Schlottmann I, Palumbo-Zerr K, Zerr P, et al. Inhibition of H3K27 histone trimethylation activates fibroblasts and induces fibrosis. Annals of the rheumatic diseases. 2013;72(4):614–20. doi: 10.1136/annrheumdis-2012-201615. [DOI] [PubMed] [Google Scholar]
- 125.Ciechomska M, O’Reilly S, Przyborski S, Oakley F, Bogunia-Kubik K, van Laar JM. Histone Demethylation and Toll-like Receptor 8-Dependent Cross-Talk in Monocytes Promotes Transdifferentiation of Fibroblasts in Systemic Sclerosis Via Fra-2. Arthritis & rheumatology (Hoboken, NJ) 2016;68(6):1493–504. doi: 10.1002/art.39602. [DOI] [PubMed] [Google Scholar]
- 126.Wang Z, Jinnin M, Nakamura K, Harada M, Kudo H, Nakayama W, et al. Long non-coding RNA TSIX is upregulated in scleroderma dermal fibroblasts and controls collagen mRNA stabilization. Experimental dermatology. 2016;25(2):131–6. doi: 10.1111/exd.12900. [DOI] [PubMed] [Google Scholar]