Mecp2 S80 phosphorylation regulates gene expression in CCl 4 -induced liver fibrosis. (A) Gene expression variances between WT and Mecp2 S80A fibrotic livers displayed as PCA plots of scaled log2-transformed transcript counts. (B) Heatmap displaying results of transcriptome sequencing carried out using 3 CCl 4 -injured livers from each of the WT and Mecp2 S80A mice; genes with greater than 2-fold change and significant to P < .01 are shown. Green indicates negative values (downregulated in Mecp2 S80A ); red, positive (up-regulated in Mecp2 S80A ); black, unchanged. (C, D) IPA was used to form a network of focused genes that are downstream targets of differentially expressed RNAs. Blue nodes show down-regulated genes in Mecp2 S80A CCl 4 -injured liver; red nodes are up-regulated. Symbol shapes signify the nature of the encoded protein (see Supplementary Figures 1 and 2 for shape information), and unshaded symbols signify genes relevant to the pathway but not differentially expressed in our data set. signif diff, significantly different.

Mecp2 S80 phosphorylation regulates gene expression in CCl 4 -induced liver fibrosis. (A) Gene expression variances between WT and Mecp2 S80A fibrotic livers displayed as PCA plots of scaled log2-transformed transcript counts. (B) Heatmap displaying results of transcriptome sequencing carried out using 3 CCl 4 -injured livers from each of the WT and Mecp2 S80A mice; genes with greater than 2-fold change and significant to P < .01 are shown. Green indicates negative values (downregulated in Mecp2 S80A ); red, positive (up-regulated in Mecp2 S80A ); black, unchanged. (C, D) IPA was used to form a network of focused genes that are downstream targets of differentially expressed RNAs. Blue nodes show down-regulated genes in Mecp2 S80A CCl 4 -injured liver; red nodes are up-regulated. Symbol shapes signify the nature of the encoded protein (see Supplementary Figures 1 and 2 for shape information), and unshaded symbols signify genes relevant to the pathway but not differentially expressed in our data set. signif diff, significantly different.

Source publication
Article
Full-text available
Background & aims: Methyl-CpG binding protein 2, MECP2, which binds to methylated regions of DNA to regulate transcription, is expressed by hepatic stellate cells (HSCs) and is required for development of liver fibrosis in mice. We investigated the effects of MECP2 deletion from HSCs on their transcriptome and of phosphorylation of MECP2 on HSC ph...

Contexts in source publication

Context 1
... These data encouraged us to examine the role of pMecp2 S80 in HSCs using primary cells isolated from digested livers of mutant Mecp2 S80A knock-in (S80A) mice. Although we were able to establish Mecp2 S80A HSC lines, they were morphologically observed to be more rounded (ie, less activated) and were slower growing than WT lines (Supplementary Figure 7); the reduced proliferative rate was confirmed by BrdU incorporation ( Figure 4B), and MTT assays were carried out across a time course of transdifferentiation ( Figure 4C). Moreover, S80A HSCs were impaired for expression of Mcm5 transcript and protein ( Figure 4D) and displayed diminished expression of CCNA2 and Brip1 ( Figure 4E). ...
Context 2
... the protective effect of S80A point mutation on fibrosis, we performed transcriptomic sequencing of CCl 4 -injured livers. Sequencing data from 2 groups were normalized and used to generate a PCA plot, which showed clear phenotypic differences between the WT and S80A ( Figure 7A). Interrogation of the sequencing data showed reduced expression of 116 genes and up-regulation of 306 genes in S80A livers compared with the WT ( Figure 7B and Supplementary Table 6). ...
Context 3
... data from 2 groups were normalized and used to generate a PCA plot, which showed clear phenotypic differences between the WT and S80A ( Figure 7A). Interrogation of the sequencing data showed reduced expression of 116 genes and up-regulation of 306 genes in S80A livers compared with the WT ( Figure 7B and Supplementary Table 6). IPA analysis of differentially expressed genes in S80A-injured livers showed similar networks to those identified in Mecp2 -/y myofibroblasts, particularly pathways involving histones H3 and H4, extracellular signal-regulated kinases 1 and 2, and RNA polymerase II ( Figure 7C and D and Supplementary Figures 1 and 2). ...
Context 4
... of the sequencing data showed reduced expression of 116 genes and up-regulation of 306 genes in S80A livers compared with the WT ( Figure 7B and Supplementary Table 6). IPA analysis of differentially expressed genes in S80A-injured livers showed similar networks to those identified in Mecp2 -/y myofibroblasts, particularly pathways involving histones H3 and H4, extracellular signal-regulated kinases 1 and 2, and RNA polymerase II ( Figure 7C and D and Supplementary Figures 1 and 2). These data suggest a role for S80 phosphorylation in controlling Mecp2-mediated regulation of chromatin structure and gene transcription as well as mitogenactivated protein kinase signaling cascades in hepatic myofibroblasts and liver fibrosis. ...

Citations

... The findings, which were inconsistent with previous reports, may be related to the small number of patients and the different study contexts. It is also possible that HA expression may be affected by several uncontrollable confounders in clinical practice, such as chronic liver complications and individual heterogeneity (Heldin et al., 2019;Moran-Salvador et al., 2019). More likely, SARS-CoV-2 promotes the accumulation of HA by activating HA synthase, thereby masking the minor HA fluctuations caused by diabetes mellitus or atherosclerosis . ...
Article
Full-text available
Objective Human identical sequences of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) promoted the coronavirus disease 2019 (COVID-19) progression by upregulating hyaluronic acid (HA) via NamiRNA-enhancer network, based on previous experimental research. This study aimed to investigate the predictive value of HA for the severity of SARS-CoV-2 infection in the post-COVID-19 era. Methods A total of 217 consecutive patients with COVID-19 were enrolled at Beijing Ditan Hospital between July 2023 and October 2023. HA levels were analyzed using biochemical detector. Logistic regression analysis was used to screen independent factors for severe COVID-19. The predictive performance of HA for severe infection was assessed by ROC curve. Furthermore, the relationship between HA levels and COVID-19 severity was investigated using multivariate logistic regression models after adjustment for potential confounders. Results According to the cut-off value of HA, COVID-19 patients were divided into HA < 90 ng/mL group (80 cases) and HA ≥ 90 ng/mL group (137 cases). High HA levels were positively associated with the severe SARS-CoV-2 infection, including elevated inflammatory indicators, severe lung involvement, prolonged clinical course, and higher incidence of respiratory failure and death (P < 0.05). Logistic regression analysis suggested that HA was an independent predictor of severe COVID-19 (OR = 4.540, 95% CI = 2.105-9.790, P < 0.001). ROC curve analysis showed that the AUC of HA for severe infection was 0.724. HA levels were significantly higher in COVID-19 cases compared to the healthy population (123.9 (82.6, 174.1) vs. 50.5 (37.8, 66.8), P < 0.001), but similar to those with non-SARS-CoV-2 lung infection (121.6 (78.5, 175.6) vs. 106.0 (66.5, 149.7), P = 0.244). We also found that the first COVID-19 infections had higher HA levels (118.8 (79.5, 174.3) vs. 85.0 (61.1, 128.8), P < 0.001) and a higher proportion of severe infection (37.1% vs. 21.3%, P = 0.043) than re-infections. However, HA expression failed to fully return to normal levels with infection recovery (204.7 (152.9, 242.2) vs. 97.0 (69.3, 137.3), P < 0.001). Conclusion HA was associated with severe SARS-CoV-2 infection and could be used as a novel serum biomarker to predict the risk of COVID-19 progression in the post-COVID-19 era.
... It was reported that phosphorylation of MeCP2 leads to HSCs proliferation and hepatic fibrogenesis. 13 DNA methyltransferases (DNMTs) promote DNA methylation by regulating gene expression. 14 The DNMTs family mainly comprises DNMT1, DNMT3A, DNMT3B and DNMT3L. ...
Article
Full-text available
Non-alcoholic fatty liver disease (NAFLD) is an extremely prevalent disease, and the presence and severity of liver fibrosis are considered one of the most important factors determining its prognosis. Hepatic stellate cells (HSCs) are essential in hepatic fibrogenesis associated with NAFLD. A number of factors underlying NAFLD pathogenesis may promote HSCs activation, leading to the development of profibrotic and proinflammatory signs. In addition, for the fibrogenic transdifferentiation of quiescent HSCs, alterations in multiple genes are necessary, where epigenetic regulation plays a defining role. Epigenetic regulation induces changes in gene activity without altering the coding sequence, and these changes are stably inherited after the factor causing the alteration has disappeared. Epigenetic modifications comprise several regulatory mechanisms, includ- ing DNA methylation, covalent histone modification, chromatin remodeling, and non-coding RNAs. Since the mechanisms underlying epigenetic regulation of HSCs fibrogenic activation are reversible and dynamic, molecular targeted therapies aimed at correcting these mechanisms provide promising prospects for novel therapeutic approaches for treating liver fibrosis associated with NAFLD.
... Under myofibroblast phenotypic transformation, methyl CpG binding protein 2 (MeCP2), with high affinity to methylated DNA, maintained the hypermethylation (one with 109 CpG and the other with 21 CpG) of PPARg promoter region and thus facilitated PPARg suppression and liver fibrosis progression 18 . Recently, Moran-Salvador et al. 19 established MeCP2 deletion and MeCP2 S80A mutated mouse model and further verified that the phosphorylation of MeCP2 at S80 site is required for HSC activation and collagen expression in the CCl 4 mouse model. Additionally, MeCP2 upregulated myofibroblast DNA replication-related genes, including Cdc7, Has2 and Dna2, and thus enhanced active HSC proliferation and hepatic fibrogenesis. ...
Article
Full-text available
Liver fibrosis, characterized by scar tissue formation, can ultimately result in liver failure. It's a major cause of morbidity and mortality globally, often associated with chronic liver diseases like hepatitis or alcoholic and non-alcoholic fatty liver diseases. However, current treatment options are limited, highlighting the urgent need for the development of new therapies. As a reversible regulatory mechanism, epigenetic modification is implicated in many biological processes, including liver fibrosis. Exploring the epigenetic mechanisms involved in liver fibrosis could provide valuable insights into developing new treatments for chronic liver diseases, although the current evidence is still controversial. This review provides a comprehensive summary of the regulatory mechanisms and critical targets of epigenetic modifications, including DNA methylation, histone modification, and RNA modification, in liver fibrotic diseases. The potential cooperation of different epigenetic modifications in promoting fibrogenesis was also highlighted. Finally, available agonists or inhibitors regulating these epigenetic mechanisms and their potential application in preventing liver fibrosis were discussed. In summary, elucidating specific druggable epigenetic targets and developing more selective and specific candidate medicines may represent a promising approach with bright prospects for the treatment of chronic liver diseases.
... In total, 82 DEGs were downregulated in patients with RTT and upregulated in patients with MDS, indicating that MECP2 is important for mRNA processing. Moran-Salvador et al. found a downregulated group of genes involved in DNA replication and cell proliferation in hepatic stellate cells of Mecp2-null mice and suggested inhibition of Mecp2 phosphorylation as a liver fibrosis treatment [61]. ...
Article
Full-text available
Background Rett syndrome (RTT) is a neurodevelopmental disorder mainly caused by mutations in the methyl-CpG-binding protein 2 gene (MECP2). MeCP2 is a multi-functional protein involved in many cellular processes, but the mechanisms by which its dysfunction causes disease are not fully understood. The duplication of the MECP2 gene causes a distinct disorder called MECP2 duplication syndrome (MDS), highlighting the importance of tightly regulating its dosage for proper cellular function. Additionally, some patients with mutations in genes other than MECP2 exhibit phenotypic similarities with RTT, indicating that these genes may also play a role in similar cellular functions. The purpose of this study was to characterise the molecular alterations in patients with RTT in order to identify potential biomarkers or therapeutic targets for this disorder. Methods We used a combination of transcriptomics (RNAseq) and proteomics (TMT mass spectrometry) to characterise the expression patterns in fibroblast cell lines from 22 patients with RTT and detected mutation in MECP2, 15 patients with MDS, 12 patients with RTT-like phenotypes and 13 healthy controls. Transcriptomics and proteomics data were used to identify differentially expressed genes at both RNA and protein levels, which were further inspected via enrichment and upstream regulator analyses and compared to find shared features in patients with RTT. Results We identified molecular alterations in cellular functions and pathways that may contribute to the disease phenotype in patients with RTT, such as deregulated cytoskeletal components, vesicular transport elements, ribosomal subunits and mRNA processing machinery. We also compared RTT expression profiles with those of MDS seeking changes in opposite directions that could lead to the identification of MeCP2 direct targets. Some of the deregulated transcripts and proteins were consistently affected in patients with RTT-like phenotypes, revealing potentially relevant molecular processes in patients with overlapping traits and different genetic aetiology. Conclusions The integration of data in a multi-omics analysis has helped to interpret the molecular consequences of MECP2 dysfunction, contributing to the characterisation of the molecular landscape in patients with RTT. The comparison with MDS provides knowledge of MeCP2 direct targets, whilst the correlation with RTT-like phenotypes highlights processes potentially contributing to the pathomechanism leading these disorders.
... It can bind to methylated or unmethylated DNA at 5′-CpG residues and recruit corepressor complexes to suppress gene expression by DNA methylation and histone modifications (Boxer et al. 2020). Several studies have also revealed that MeCP2 promotes hepatic and pulmonary fibrosis (Hu et al. 2011;Moran-Salvador et al. 2019). Thus, MeCP2 has been proposed as a potential target to attenuate TGFβ-induced EndMT and organ fibrosis (He et al. 2018). ...
Article
Full-text available
Endothelial-to-mesenchymal transition (EndMT), the process by which endothelial cells lose their characteristics and acquire mesenchymal phenotypes, participates in the pathogenic mechanism of idiopathic pulmonary fibrosis. Recently, exosomes derived from human umbilical cord mesenchymal stem cells (hucMSC-Exos) has been introduced as a promising treatment in organ fibrosis. This study aimed to explore the effects as well as the molecular mechanism for hucMSC-Exo in pulmonary fibrosis. The intravenous administration of hucMSC-Exos alleviated bleomycin-induced pulmonary fibrosis in vivo. Moreover, hucMSC-Exos elevated miR-218 expression and restored endothelial properties weakened by TGF-β in endothelial cells. Knockdown of miR-218 partially abrogated the inhibition effect of hucMSC-Exos on EndMT. Our mechanistic study further demonstrated that MeCP2 was the direct target of miR-218. Overexpressing MeCP2 aggravated EndMT and caused increased CpG islands methylation at BMP2 promoter, which lead to BMP2 post-transcriptional gene silence. Transfection of miR-218 mimic increased BMP2 expression as well, which was downregulated by overexpression of MeCP2. Taken together, these findings indicate exosomal miR-218 derived from hucMSCs may possess anti-fibrotic properties and inhibit EndMT through MeCP2/BMP2 pathway, providing a new avenue of preventive application in pulmonary fibrosis. Graphical abstract
... [39] HSCs with knockdown or knockout of HAS2 show reduced collagen production, HSC migration, and proliferation in vitro. [4,40] Also, HAS2 is crucial for HA production, cancer growth in cholangiocarcinoma, and liver metastasis caused by colorectal and pancreatic cancers. [41,42] These findings indicate that HAS2 is the isozyme responsible for HA production in liver fibrosis and liver malignancy; HAS2 and HA are primarily expressed in HSCs; and HAS2 and HA contribute to HSC activation, liver fibrosis, and fibrosis-mediated cancer progression. ...
... HAS2 expression is also regulated epigenetically. [40] For example, an epigenetic regulator methyl-CpG binding protein 2 regulates the expression of profibrogenic genes through phosphorylation at serine 80 in HSCs. HAS2 is one of the profibrogenic genes regulated by methyl-CpG binding protein 2. A post-transcriptional regulation of HAS2 was also reported. ...
Article
Full-text available
Hyaluronan (HA), also known as hyaluronic acid, is a glycosaminoglycan that is a critical component of the extracellular matrix (ECM). Production and deposition of ECM is a wound-healing response that occurs during chronic liver disease, such as cirrhosis. ECM production is a sign of the disease progression of fibrosis. Indeed, the accumulation of HA in the liver and elevated serum HA levels are used as biomarkers of cirrhosis. However, recent studies also suggest that the ECM, and HA in particular, as a functional signaling molecule, facilitates disease progression and regulation. The systemic and local levels of HA are regulated by de novo synthesis, cleavage, endocytosis, and degradation of HA, and the molecular mass of HA influences its pathophysiological effects. However, the regulatory mechanisms of HA synthesis and catabolism and the functional role of HA are still poorly understood in liver fibrosis. This review summarizes the role of HA in liver fibrosis at molecular levels as well as its clinical implications and discusses the potential therapeutic uses of targeting HA in liver fibrosis.
... Of note, MeCP2 was reported as a regulator of miRs/genes involved in liver fibrosis. [42][43][44] Regarding results on ESR1, we speculate that its effect on miR regulation may also depend on estrogen level in the cells. Because LX2 is a male cell line, the regulation of ESR1 on sex-specific DEmiRs may not be fully characterized by manipulation of gene targets alone. ...
Article
Full-text available
Liver cirrhosis represents an advanced stage of chronic liver disease and is associated with significant morbidity, mortality, and risk of cancer development. While sex disparity of liver diseases has been observed, understanding at a genetic level awaits more thorough investigation. In this study, we performed a sex‐specific analysis of the microRNA (miR) profiles in hepatitis B virus (HBV)–associated cirrhosis by small RNA‐sequencing using clinical tissue samples. Potential associated signaling pathways, downstream gene targets, and upstream regulators were highlighted by computational prediction analyses based on the differentially expressed miRs (DEmiRs). From our results, deregulation of miRs in cirrhosis showed a marked difference between males and females by the degree and pattern. Sixty‐five (64 up‐regulated, 1 down‐regulated) and 12 (6 up‐regulated, 6 down‐regulated) DEmiRs were found in males and females, respectively, when compared with their respective control group. A number of DEmiRs were only observed in one sex but not the other. In addition, 26 DEmiRs were identified between cirrhosis female and cirrhosis male groups. Fatty acid biosynthesis pathway, extracellular matrix–receptor interaction, p53 signaling, Hippo signaling, tumor necrosis factor signaling, the forkhead box O signaling, as well as gene targets ribosomal protein S27 like, methyl CpG binding protein 2, and estrogen receptor 1, may contribute to the pathogenesis and biological behavior of cirrhosis in a sex‐specific manner. Analysis of the Cancer Genome Atlas data set suggested a role of sex‐specific DEmiRs in multistep hepatocarcinogenesis. Conclusion: Our findings illustrate that miR profiles in HBV‐associated cirrhosis are distinct between the males and females and suggest a potential role of sex‐specific biomarkers and molecular mechanisms in disease development and progression.
... HSCs go through myofibroblast transdifferentiation to acquire a migratory, myofibroblast-like phenotype is the key step in liver fibrosis. This is a multifactorial process involving excessive proliferation, apoptotic resistance, dysregulated metabolism, and remodeling of ECM [11]. All these fibrosis-inducing pathophysiological disorders demonstrate an intimate association with abnormality in various kinds of ncRNAs (e.g., miRNA, lncRNA, and circRNA) [12]. ...
Article
Full-text available
Long noncoding RNAs (lncRNAs) are key regulators of hepatic stellate cells (HSCs), yet the role of upregulated lncRNA-NONRATT013819.2 in activated HSCs remains uncertain. In this study, the effects of NONRATT013819.2 on proliferation, apoptosis, migration, and contraction of transforming growth factor (TGF)-β1-induced HSCs were investigated. The mechanisms of NONRATT013819.2 on the activated HSCs were explored by loss-of-function of NONRATT013819.2 and gain-of-function of the target gene. Here, TGF-β1 treatment resulted in a gradual increase in the expression of cytoskeleton markers (collagen, α-SMA, and TIMP1), NONRATT013819.2, miR24-3p, and lysyl oxidase (Lox) over time in HSCs. NONRATT013819.2 acted as a sponge of miR24-3p to competitively abolish the inhibition of the lox gene in HSCs. Silencing of NONRATT013819.2 suppressed the expression of cytoskeleton markers, proliferation, and the proportion of cells that entered the S-phase, and promoted apoptosis in TGF-β1-activated HSCs. These effects were reversed when lox overexpression was introduced simultaneously. Similarly, silencing of NONRATT013819.2 also blocked ECM reconstruction, while recused by lox overexpression in TGF-β1-activated HSCs. In conclusion, upregulation of NONRATT013819.2 promotes the myofibroblastic transition by competitively binding miR24-3p to release lox in HSCs. Therefore, targeted therapy of NONRATT013819.2 may have the potential for liver fibrosis.
... 53 Peroxisome proliferator activated receptor-c (PPARG) is one of many genes that is differentially methylated during the process of HSC transdifferentiation. MECP2 promotes the methylation and repression of PPARG and inhibition of MECP2 was associated with reduced fibrosis in murine models. 54,55 Likewise, the histone methyltransferase G9a activity, alongside DNMT1, was linked to the fibrogenic activation of HSCs in patients with chronic liver injury including those with ARLD. 56 Intriguingly, the use of the novel dual G9a/DNMT1 inhibitor CM272 reduced the burden of fibrosis in mouse models. ...
Article
Full-text available
Alcohol related liver disease (ARLD) is a primary cause of chronic liver disease in the United States. Despite the advancements made in the diagnosis and management of ARLD, it remains a major public health problem with significant morbidity and mortality, which emphasizes the need for adopting novel approaches to the study of ARLD and its complications. Epigenetic changes are increasingly being recognized as contributing to the pathogenesis of multiple disease states. Harnessing the power of innovative technologies for the study of epigenetics (e.g., next generation sequencing, DNA methylation assays, histone modification profiling and computational techniques like machine learning) resulted in a seismic shift in our understanding of the pathophysiology of ARLD. Knowledge of these techniques and advancements is of paramount importance for the practicing hepatologist and researchers alike. Accordingly, in this review article we will summarize the current knowledge about alcohol induced epigenetic alterations in the context of ARLD, including but not limited to, DNA hyper/hypo methylation, histone modifications, changes in noncoding RNA, three dimensional (3D) chromatin architecture and promotor-enhancer interactions. Additionally, we will discuss the state-of-the-art techniques used in the study of ARLD (e.g., single cell sequencing). We will also highlight the epigenetic regulation of chemokines and their proinflammatory role in the context of ARLD. Lastly, we will feature the clinical applications of epigenetics in the diagnosis and management of ARLD.
... Furthermore, MeCP2 induces the expression of the H3K4 methyltransferase ASH1 (absent small and homeotic disks protein 1), which opposes the action of PPARγ by positively regulating the expression of profibrogenic genes [139,140]. In line with these results, miR-132 was recently linked to human NAFLD [141], and strategies targeting MeCP2 and EZH2 have succeeded in decreasing fibrogenic markers characteristics [142,143]. Additionally, a novel mechanism was shown to promote hepatic lipogenesis through the lncRNA-H19/mi-130a/PPARγ axis [126], becoming a potential target to treat NAFLD. ...
Article
Full-text available
Peroxisome proliferator-activated receptors (PPARs) are ligand-modulated nuclear receptors that play pivotal roles in nutrient sensing, metabolism, and lipid-related processes. Correct control of their target genes requires tight regulation of the expression of different PPAR isoforms in each tissue, and the dysregulation of PPAR-dependent transcriptional programs is linked to disorders, such as metabolic and immune diseases or cancer. Several PPAR regulators and PPAR-regulated factors are epigenetic effectors, including non-coding RNAs, epigenetic enzymes, histone modifiers, and DNA methyltransferases. In this review, we examine advances in PPARα and PPARγ-related epigenetic regulation in metabolic disorders, including obesity and diabetes, immune disorders, such as sclerosis and lupus, and a variety of cancers, providing new insights into the possible therapeutic exploitation of PPAR epigenetic modulation.