Open Access

Regulatory long non‑coding RNAs of hepatic stellate cells in liver fibrosis (Review)

  • Authors:
    • Zhengjie Wu
    • Shunmei Huang
    • Xiaoqin Zheng
    • Silan Gu
    • Qiaomai Xu
    • Yiwen Gong
    • Jiaying Zhang
    • Bin Fu
    • Lingling Tang
  • View Affiliations

  • Published online on: February 11, 2021     https://doi.org/10.3892/etm.2021.9782
  • Article Number: 351
  • Copyright: © Wu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Liver fibrosis (LF) is a continuous wound healing process caused by numerous chronic hepatic diseases and poses a major threat to human health. Activation of hepatic stellate cells (HSCs) is a critical event in the development of hepatic fibrosis. Long non‑coding RNAs (lncRNAs) that are involved in HSC activation, participate in the development of LF and are likely to be therapeutic targets for LF. In the present review, the cellular signaling pathways of LF with respect to HSCs were discussed. In particular, this present review highlighted the current knowledge on the role of lncRNAs in activating or inhibiting LF, revealing lncRNAs that are likely to be biomarkers or therapeutic targets for LF. Additional studies should be performed to elucidate the potential of lncRNAs in the diagnosis and prognosis of LF and to provide novel therapeutic approaches for the reversion of LF.

1. Introduction

Liver fibrosis (LF) results from impaired wound healing caused by acute or chronic exposure to detrimental factors, including alcohol, viral diseases, drugs, cholestasis, toxins and metabolic disorders (1,2). Slight or transient fibrosis is necessary for wound healing and maintenance of tissue architecture integrity (3). Fibrosis is a reversible process that may be halted by removing the harmful stimulus (4,5). However, severe or advanced fibrosis is characterized by abnormal connective tissue hyperplasia and extracellular matrix (ECM) protein deposition (6), leading to liver structural destruction and even organ failure (7). The accumulation of ECM proteins results in the distortion of hepatic architecture due to scar formation, along with the appearance of regenerating hepatocyte nodules, which define cirrhosis (8). Cirrhosis is characterized by hepatocellular dysfunction, portal hypertension, hepatocellular carcinoma (HCC) and eventual liver failure (9,10).

Liver fibrogenesis is initiated by hepatic stellate cells (HSCs). Under specific conditions, quiescent HSCs are transformed into myofibroblasts (MFs) to generate ECM proteins, tissue inhibitors of metalloproteinases (TIMPs) and matrix metalloproteinases (MMPs) (11-14). HSC activation involves a systemic and complex pathological process involving multiple cytokines and multiple cellular signaling pathways (15).

The healthy liver has a strong regenerative potential due to the unlimited proliferative potential of cholangiocytes and hepatocytes (HCs). Hepatic stem/progenitor cells (HpSCs) are positioned within the canals of Hering (16). These cells are quiescent in the healthy liver but may be activated in response to liver injury by proliferating and differentiating towards cholangiocytes and HCs (17). In injured tissue, activated HSCs, macrophages and MFs produce a variety of signals through signaling pathways including the Wnt and Notch pathways to drive HpSC proliferation and differentiation (18). HpSCs are also able to activate stellate cells through signaling pathways, including the Hedgehog (Hh) pathway, resulting in the release of various types of matrix components during liver regeneration (19).

Studies have confirmed that fibrosis is regulated through the expression of various genes (20). As key regulators of multiple biological processes, long non-coding RNAs (lncRNAs), commonly defined as RNAs longer than 200 nucleotides without any protein-coding capacity, have attracted much research interest (21). Although the classification system of lncRNAs is currently incomplete, they are generally divided into two broad types according to their position relative to protein-coding genes: i) Intergenic lncRNAs and ii) coding gene-overlapping lncRNAs (22). lncRNAs regulate gene expression and protein synthesis through multiple mechanisms (23) and are considered to include ~30,000 different transcripts, accounting for a large portion of the non-coding transcriptome in humans (24). Unlike mRNAs, most lncRNAs are expressed at low levels, cell type-specific, associated with a high number of coding genes (24) and are present at specific positions within the nucleus (25). lncRNAs are essential in the regulation of cell migration, apoptosis, differentiation and proliferation processes (26,27). Various lncRNAs have been confirmed to be involved in multiple diseases and certain lncRNAs have been identified as disease biomarkers (28). In addition, recent studies have revealed that lncRNAs are linked to the complex pathophysiological changes observed in LF. Although numerous lncRNAs have been identified and proposed as promising targets for anti-fibrosis therapies, the underlying mechanisms of the functions of lncRNAs in LF have remained elusive.

In the present review, the role of lncRNAs in modulating cellular signaling pathways in LF was explored. The potential utility of lncRNAs as non-invasive biomarkers and novel therapeutic targets for LF was also proposed.

2. Transforming growth factor-β (TGF-β)/Smad signaling and lncRNAs

TGF-β is generally considered the core driving factor behind LF (29-31). Mammals have three types of TGF-β (β1, β2 and β3). TGF-β1 acts as an important regulator of fibrogenesis, notably in inflammation-induced LF (32). TGF-β1 binding to TGF-β receptor (TGFβR)I and TGFβRII triggers the phosphorylation of downstream Smad proteins (particularly Smad3) and contributes to the transcription of type I and type III collagen mRNAs (33,34). TGF-β1 increases TIMP expression and decreases MMP levels, thereby inhibiting ECM degradation. In addition, TGF-β1 induces MF production by promoting epithelial-mesenchymal transition (EMT). TGF-β1 promotes matrix formation through Smad3-dependent as well as Smad3-independent mechanisms (35).

lncRNA-activated by TGF-β (lncRNA-ATB) is an important regulator of the TGF-β/Smad signaling pathway. Studies have reported that lncRNA-ATB, Smad2 and TGFβRII share a common microRNA (miRNA) response element (MRE) for miRNA (miR)-425-5p. lncRNA-ATB was indicated to induce the expression of Smad2 and TGF-βRII by inhibiting the expression of endogenous miR-425-5p in hepatitis C virus (HCV)-induced hepatic fibrosis in a study on hepatic stellate LX-2 cells treated with hepatoblastoma HepG2 cells carrying the HCV core protein. Consequently, lncRNA-ATB caused hepatic fibrosis by enhancing collagen I synthesis and stimulating HSCs through competitive binding to miR-425-5p (36).

LF-associated lncRNA1 (lnc-LFAR1) was indicated to facilitate the interaction between Smad2/3 and TGFβRI and promote Smad2/3 phosphorylation in carbon tetrachloride (CCl4)/bile duct ligation (BDL)-induced LF. In addition, lnc-LFAR1 is able to directly bind to Smad2/3. On this basis, the TGFβ1/Smad2/3/lnc-LFAR1 signaling pathway creates an active feedback loop that enhances Smad2/3 functions in hepatic fibrosis (37).

HOXA distal transcript antisense RNA (HOTTIP) has been implicated in liver fibrogenesis (38). Li et al (39) determined that HOTTIP was upregulated in mice with hepatic fibrosis and that inhibition of HOTTIP by adenoviral delivery of short hairpin RNA-HOTTIP markedly reduced LF (39). miR-148a participates in the initiation and progression of HCC in the presence of LF and HOTTIP inhibits miR-148a expression (40). miR-148a may regulate TGFβRI/TGFβRII, subsequently decreasing their expression levels, in human and mouse HSCs. Collectively, these results suggest that HOTTIP may promote LF by downregulating miR-148a and upregulating TGFβRI and TGFβRII.

H19 is a maternally inherited gene (41) and is overexpressed in human hepatic fibrosis specimens, as well in the livers of mice with CCl4/BDL-induced fibrosis (42). H19 functions as a competing endogenous RNA (ceRNA) by sponging miR-148a and maintaining the expression levels of ubiquitin-specific protease 4 (USP4), a key miR-148a target that stabilizes TGFβRI and promotes TGF-β signalling (43). The H19/miR-148a/USP4 axis activates hepatic fibrosis through the TGF-β pathway, indicating that H19 may be a therapeutic target for fibrosis (44).

The lncRNA Gm5091 significantly negatively regulates HSCs in mice with alcoholic hepatic fibrosis (AHF) (45). Zhou et al (45) reported that Gm5091 downregulates cell migration, collagen I expression and HSC activation marker expression, including Desmin and α-smooth muscle actin. In addition, data based on a bioinformatic analysis revealed that the Gm5091 sequence contains binding sites for miR-24, miR-23b and miR-27b. Full-length Gm5091 decreases the expression levels of miR-27b/23b/24(45). In addition, miR-27, miR-24 and miR-23 are positive regulators of HSC proliferation and differentiation, as they activate TGF-β and Smad4 in mice with LF (46,47). As a ceRNA, Gm5091 sponges miR-27b/23b/24, which alleviates liver injury and the progression of AHF in mice by inhibiting HSC activation.

Nuclear paraspeckle assembly transcript 1 (NEAT1) is critical for the formation of paraspeckles (48) and hence to the initiation of tumors. It is highly expressed in activated HSCs and liver tissue of mice with CCl4-induced LF. In human fibrotic liver samples, upregulated expression levels of NEAT1 are positively correlated with fibrosis markers (49). In a mouse model of CCl4-induced fibrosis, NEAT1 overexpression promoted HSC activation in vivo, and similar results were obtained in vitro (50). A previous study suggested that miR-122 inhibits HSC activation and the expression of fibrosis-associated genes induced by TGF-β (51). Stimulated expression of Kruppel-like factor 6 (KLF6), an immediate-early gene in LF, induces the expression of TGFβRI and TGFβRII in activated HSCs (52). Furthermore, miR-122 targets NEAT1 as well as KLF6(53). Collectively, these results suggest that NEAT1 competitively binds to miR-122 and regulates KLF6 expression in hepatic fibrosis, indicating that the NEAT1/miR-122/KLF6 axis promotes HSC activation.

The lncRNA ENSMUST00000158992 (SCARNA10) is upregulated in liver tissues of fibrotic mice as well as in the serum and liver tissue of humans with advanced LF (54). Several studies have indicated that SCARNA10 is a positive regulator of LF, as it induces HC apoptosis and HSC activation (55,56). Mechanistically, SCARNA10 functions as a mediator of LF by inhibiting the binding of polycomb repressive complex 2 to the promoters of genes involved in the TGF-β pathway, thereby promoting the transcription of these genes.

3. Hh signaling and lncRNAs

The Hh signaling pathway is a morphogenic pathway that has multiple roles in cell proliferation, apoptosis, migration and differentiation. It was first reported in Drosophila by Nüsslein-Volhard and Wieschaus (57) in 1980. The Hh pathway comprises Glioblastoma (GLI) family transcription factors (GLI1, GLI2, GLI3), Smoothened (SMO), sonic Hh and Patched 1 (PTCH1) (58) and is driven by PTCH receptors that are activated by Hh ligands, which abolish the inhibitory effect of PTCH1 on SMO. In turn, SMO transduces Hh signals to regulate gene expression via GLI transcription factors (59,60). Normal adult HCs generally do not produce Hh ligands but hepatic synthesis of Hh ligands is increased in liver injury (61). In addition, several resident liver cells, including HCs, HSCs, cholangiocytes, macrophages, natural killer T cells and liver sinusoidal endothelial cells, are able to produce Hh ligands (62,63). Stimuli that contribute to liver regeneration/remodeling promote the expression of Hh ligands in the liver. HSCs become highly activated by Hh ligands, which enhances their fibrogenic and proliferative capabilities (64-66). Activation of the Hh pathway is influenced by liver regeneration, hepatic accumulation of inflammatory cells, liver fibrogenesis and vascular remodelling (67). In addition, the Hh pathway activity is positively associated with the fibrosis stage (68). Data from Sicklick et al (69) indicate that inactivation of Hh signaling in HSCs inhibits HSC activation in vitro. Approaches that may block the Hh pathway may not only reduce HSC activation and fibrosis but also prevent the accumulation of hepatic progenitor cells (70). Furthermore, EMT is a key event in HSC activation and is regulated by the Hh pathway (71,72).

Plasmacytoma variant translocation 1 (PVT1) promotes HSC activation through the Hh pathway and EMT process (73). PTCH1 is a negative modulator of Hh signalling (58). PVT1 knockdown increases the expression of PTCH1. Inhibition of PTCH1 during liver fibrogenesis results in PTCH1 methylation, whereas silencing of PTCH1 expression promotes activation of the Hh pathway in CCl4-induced LF (74). Furthermore, PVT1 inhibits miR-152 through a post-transcriptional mechanism, while miR-152 promotes hypomethylation of PTCH1 by suppressing its direct target DNA methyltransferase 1(75). Demethylation of PTCH1 by miR-152 modulates the effect of PVT1 inhibition on PTCH1 levels and treatment with a miR-152 antagonist abolishes these changes. Therefore, PVT1 inhibits PTCH1 through competitive binding with miR-152 and promotes EMT in hepatic fibrosis.

The lncRNA-maternally expressed gene 3 (MEG3) was indicated to be downregulated in hepatic fibrosis in vitro and in vivo, and its overexpression alleviates fibrogenesis (76,77). Previous studies have reported that MEG3 suppresses hepatic fibrosis via p53(78). In particular, overexpression of MEG3 inhibits HSC activation by promoting the EMT process (79). In addition, deletion of the SMO binding site in MEG3 fails to block the effects of MEG3 on the EMT process and GLI3 in mice treated with CCl4 (76). miR-212 is significantly downregulated in MEG3-overexpressing cells and is able to target PTCH1. Furthermore, MEG3 induces Hh pathway activation by sponging miR-212, promoting PTCH1 expression and decreasing SMO expression.

4. Wnt/β-catenin signaling and lncRNAs

Wnt signaling modulates cellular apoptosis, proliferation and differentiation. Wnt proteins are 350-400 amino acids long with a conserved cysteine-rich binding domain containing 23-24 cysteine residues (80). Of note, two cell surface receptor families participate in the reception and transduction of Wnt signals: The low-density lipoprotein receptor-related protein (LRP) family and members of the Frizzled (Fz) gene family (81). When Wnt binds to its receptor, either Fz or a complex formed by Fz and LRP5/6, a signal is transduced to the cytoplasmic phosphoprotein disheveled (82). Mammals have three types of Dsh proteins (Dsh-3, Dsh-2 and Dsh-1) (83). Wnt signaling is divided into three independent pathways according to the affected Dsh protein: The canonical ‘Wnt/β-catenin’ pathway, the ‘Wnt/polarity’ pathway (also called the ‘planar cell polarity’ pathway) and the ‘Wnt/Ca2+’ pathway (84-86). In these three pathways, Dsh is a key transducer of the Wnt signal.

β-catenin is a major component of the canonical Wnt pathway (87-89). β-catenin forms a subunit of the cadherin protein complex (90). Previous studies have indicated that β-catenin is involved in fibrotic diseases (91,92). In the absence of Wnt, β-catenin is targeted for degradation by a multiprotein degradation complex. Wnt signaling antagonizes the degradation complex, leading to β-catenin accumulation and target gene activation (93).

The Wnt pathway has dual modulatory effects on HSCs (94). Depending on the specific conditions, Wnt may either activate or inhibit β-catenin. Abnormal activation of Wnt/β-catenin signaling aggravates fibrogenesis. Small interfering RNA-mediated β-catenin knockdown suppresses cell proliferation and decreases the expression levels of collagen I and III, resulting in HSC apoptosis in vitro (95). PRI-724 is a selective inhibitor of the cAMP-response element-binding protein-binding protein (CBP)/β-catenin interaction, activation of HSCs by PRI-724 has been indicated to reduce hepatic fibrogenesis in mice (96). Activation of the canonical Wnt/β-catenin pathway is necessary to sustain the quiescence of HSCs in vitro (97). Roof plate-specific spondin proteins, which stimulate the Wnt pathway, have been reported to inhibit HSC activation, thereby compromising dickkopf WNT signaling pathway inhibitor 1 signaling (98). Non-canonical Wnt pathway signaling activates HSCs, as supported by the observation that overexpression of Wnt5a was activated in rat HSCs, indicating that cellular signaling involving phosphoprotein dishevelled occurs (99). On the other hand, a natural Wnt5a inhibitor inhibited HSC activation by inducing the expression of secreted Fz-related protein 5(100).

lncRNA-ATB activates EMT and promotes tumor metastasis (101), and its expression is positively correlated with liver cirrhosis in patients with HCC (102). Evidence indicates that lncRNA-ATB competitively binds to miR-200 family members (103). Furthermore, β-catenin was reported to be regulated by miR-200a (104,105). Collectively, these results suggest that lncRNA-ATB upregulates β-catenin expression by inhibiting miR-200a, resulting in collagen Ι synthesis and HSC activation in HCV-associated fibrosis in humans (106).

Long intergenic non-coding RNA-p21 (lincRNA-p21) inhibits Wnt/β-catenin signaling, which mediates the effects of salvianolic acid B (Sal B) on HSC activation. The inhibitory effects of Sal B on Wnt/β-catenin signaling are abolished by lincRNA-p21 suppression. lincRNA-p21 regulated by miR-17-5p is an inhibitor of miR-17-5p. Collectively, these results indicate that lincRNA-p21 inhibits HSC activation through the miR-17-5p/Wnt/β-catenin axis (107).

Small nuclear RNA host gene 7 (SNHG7) has been reported to be an oncogene in various cancer types. Yu et al (108) reported that the expression levels of SNHG7 are upregulated in the liver tissue of CCl4 mice and that silencing of SNHG7 inhibits HSCs. Furthermore, SNHG7 is able to regulate miR-378a-3p. Downregulation of miR-378a-3p reduces the effects of SNHG7 loss on HSC activation (109). SNHG7 enhances Wnt/β-catenin signaling, leading to LF, characterized by a decline in the phosphorylated β-catenin level and enhancement of T cell factor activity (110). SNHG7-mediated activation of the Wnt/β-catenin pathway is regulated by miR-378a-regulated disheveled segment polarity protein 2 (DVL2). Therefore, miR-378a-3p is a regulator of DVL2. DVL2 inhibition abolishes SNHG7-induced HSC activation. Collectively, these results indicate that SNHG7 inhibits miR-378a-3p and moderates its effects on DVL2, leading to enhanced Wnt/β-catenin signaling and thereby promoting hepatic fibrosis.

5. NF-κB signaling and lncRNAs

The NF-κB pathway has a crucial role in innate and adaptive immunity. NF-κB is a eukaryotic transcription factor that exists in almost all cell types and is involved in various liver pathologies (111). The survival and activation of HSCs and hepatic MFs is modulated by NF-κB (112). In 1986, NF-κB was discovered to regulate immunoglobulin κ light chain expression in B cells (113). NF-κB dimers are sequestered in the cytoplasm by inhibitor of NF-κB proteins in most resting cells (114). NF-κB activation is associated with at least 2 signal transduction routes named the canonical and non-canonical pathways (115).

NF-κB regulates LF mainly by modulating physiological responses in HSCs (116). Activation of NF-κB in HCs confers protection by limiting apoptosis and facilitating regeneration through stimulation of HC proliferation (117). NF-κB is activated in Kupffer cells during liver injury, which further induces HSC activation and liver fibrogenesis (118,119). Further studies have indicated that NF-κB agonists, including captopril, thalidomide, silymarin and sulfasalazine, enhance hepatic MF apoptosis and antifibrotic activities in normal liver tissue (120-122). In addition, HSCs are direct in vivo targets of regulators that activate NF-κB, including lipopolysaccharide (LPS) (123). However, prolonged accumulation of NF-κB in liver cells promotes chronic inflammation and HSC transdifferentiation into scar-forming hepatic MFs (124,125). By contrast, inhibition of NF-κB in Kupffer cells alleviates hepatic fibrosis (126).

In fibrotic livers and activated HSCs, the lncRNA taurine upregulated gene 1 (TUG1) is highly expressed, unlike in normal HCs. miR-29b has been confirmed as a TUG1-targeting miRNA (127). Previous research has demonstrated that miR-29b inhibits HSC activation and ameliorates CCl4/BDL-induced LF, as well as human advanced hepatic fibrosis (128). Treatment with a miR-29b mimic eliminates the effects of TUG1 overexpression on cellular physiological responses and inactivation of Janus kinase/STAT and NF-κB signaling in LPS-pretreated H9c2 cells (127). miR-29b downregulates fibrogenic genes associated with the NF-kB and TGF-β pathways in HSCs (129,130). Murine miR-29b suppresses the expression of collagen in HSCs and is downregulated in activated HSCs in a manner dependent on NF-kB and TGF-β signalling (131). Thus, TUG1 is a positive regulator of profibrogenic gene expression in HSCs, as it downregulates miR-29b in HSCs (132).

6. Notch signaling and lncRNAs

The Notch signaling pathway is an elementary and highly conserved pathway associated with liver development, physiology and pathophysiology (133,134). Early studies based on C. elegans and Drosophila genetic models identified a gene locus that was correlated with the phenotype of a mutant fly with a wing indentation (18,135). This locus was indicated to participate in cell fate processes during Drosophila embryogenesis and was later named Notch. The Notch pathway consists of receptors (Notch1-Notch4), ligands (δ-like 1, 3 and 4, as well as Jagged 1 and 2), transcriptional complex components and downstream genes, including hairy-enhancer of split (Hes)-related with YRPW motif (Hey) and Hes (136,137). Notch signaling promotes LF by regulating the inflammatory response and the function of macrophages (138,139). Xie et al (140) reported that Notch-Hh crosstalk influences the pathogenesis of cirrhosis by regulating MFs/HSCs through EMT in vitro and in vivo. In a mouse model of liver steatosis and LF, sustained Notch activation induces hepatic fibrosis in the presence of high lipid levels and inhibition of Notch signaling or a reduction in liver fat content may ameliorate hepatic fibrosis (141,142).

In mouse models of liver fibrogenesis, the protein and mRNA levels of Notch2, Notch3, Hes1 and Hey2 are increased in lnc-LFAR1-overexpressing HSCs, while downregulation of lnc-LFAR1 reverses these effects (37). Hes is considered the prototype Notch target gene and encodes a basic helix-loop-helix inhibitory transcription factor involved in the self-renewal of target cells by inhibiting differentiation (143,144). Furthermore, lentivirus-mediated knockdown of lnc-LFAR1 reduced the expression levels of Hey2, Notch2 and Notch3. Hes1 inhibits CCl4/BDL-induced expression of these genes; therefore, lnc-LFAR1 may activate HSCs and subsequently accelerate LF through modulation of Notch signaling.

7. Other signaling pathways

Further signaling pathways are involved in LF. Homeobox transcript antisense RNA (HOTAIR) expression is upregulated in HSCs during LF. HOTAIR modulates PTEN levels and contributes to the activation of the ERK and AKT pathways through miR-29b (145). In addition, the lncRNA growth arrest-specific transcript 5 inhibits LF by targeting miR-23a through the PTEN/PI3K/Akt signaling pathway in a rat model of CCl4-induced hepatic fibrosis (146). Future research is expected to focus increasingly on the association of lncRNAs with LF.

8. Conclusion

Regeneration of damaged mature liver tissue is driven by multiple signaling pathways, including the TGF-β/Smad, Hh, Wnt, NF-κB and Notch pathways. A summary is provided in Table I and Fig. 1. The complex but delicate networks interconnecting these molecular signals regulate cellular proliferation, differentiation and apoptosis and thus the pathological process of fibrosis. Due to the development of high-throughput sequencing technologies, numerous lncRNAs have been identified. These lncRNAs may act on oncogenes or tumor suppressors and certain lncRNAs have been well characterized and proven to be associated with LF. In the present review, these signals and intracellular events were summarized that independently or cooperatively drive HSC activation. The roles and possible mechanisms of action of selected lncRNAs in LF were also reviewed. The potential utility of lncRNAs as therapeutic agents and biomarkers is promising, although the exact mechanisms of action behind most lncRNAs remain elusive. Given the numerous potential therapeutic anti-fibrosis strategies targeting factors that promote fibrosis, combination therapies including these lncRNAs may produce improved clinical outcomes. However, additional functions and regulatory mechanisms of action of these lncRNAs require further study prior to their use as clinically applicable biomolecules.

Table I

lncRNAs regulating the signaling pathways in LF.

Table I

lncRNAs regulating the signaling pathways in LF.

lncRNAExperiment typeSignaling pathwayFunction(Refs.)
lncRNA-ATBIn vitroTGF-β/Smad signalingPromoting HSC activation(33)
lnc-LFAR1In vitro and in vivoTGF-β/Smad signaling, Notch signalingPromoting HSC activation(34)
HOTTIPIn vitro and in vivoTGF-β/Smad signalingPromoting HSC activation(36)
H19In vitro and in vivoTGF-β/Smad signalingPromoting HSC activation(39)
lncRNA Gm5091In vitro and in vivoTGF-β/Smad signalingInhibiting HSC activation(42)
NEAT1In vitro and in vivoTGF-β/Smad signalingPromoting HSC activation(47)
PVT1In vitro and in vivoHedgehog signalingPromoting HSC activation(66)
MEG3In vitro and in vivoHedgehog signalingInhibiting HSC activation(67)
lncRNA-ATBIn vitroWnt/β-catenin signalingPromoting HSC activation(94)
lincRNA-p21In vitroWnt/β-catenin signalingInhibiting HSC activation(95)
SNHG7In vitro and in vivoWnt/β-catenin signalingPromoting HSC activation(96)
TUG1In vitro and in vivoNF-κB signalingPromoting HSC activation(110)

[i] LF, liver fibrosis; HOTTIP, HOXA distal transcript antisense RNA; HSC, hepatocellular carcinoma; lnc-LFAR1, LF-associated lncRNA1; lincRNA-p21, long intergenic non-coding RNA-p21; lnc/lncRNA, long non-coding RNA; lncRNA-ATB, lncRNA-activated by TGF-β; MEG3, maternally expressed gene 3; NEAT1, encoding nuclear paraspeckle assembly transcript 1; PVT1, plasmacytoma variant translocation 1; SNHG7, small nuclear RNA host gene 7; TGF-β, transforming growth factor-β; TUG1, taurine upregulated gene 1.

Acknowledgements

Not applicable.

Funding

Funding: This research was funded by the National Science and Technology Major Project (no. 2017ZX10204401).

Availability of data and materials

Not applicable.

Authors' contributions

ZW, SH and LT designed the article. ZW, SH, XZ and SG wrote the first draft of the manuscript. QX, YG, JZ and BF reviewed the literature. LT critically revised the manuscript. All authors read and approved the final version of the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Friedman SL: Liver fibrosis-from bench to bedside. J Hepatol. 38 (Suppl 1):S38–S53. 2003.PubMed/NCBI View Article : Google Scholar

2 

Hernandez-Gea V and Friedman SL: Pathogenesis of liver fibrosis. Annu Rev Pathol. 6:425–456. 2011.PubMed/NCBI View Article : Google Scholar

3 

Cao L, Nicosia J, Larouche J, Zhang Y, Bachman H, Brown AC, Holmgren L and Barker TH: Detection of an integrin-binding mechanoswitch within fibronectin during tissue formation and fibrosis. ACS Nano. 11:7110–7117. 2017.PubMed/NCBI View Article : Google Scholar

4 

Kong D, Zhang F, Zhang Z, Lu Y and Zheng S: Clearance of activated stellate cells for hepatic fibrosis regression: Molecular basis and translational potential. Biomed Pharmacother. 67:246–250. 2013.PubMed/NCBI View Article : Google Scholar

5 

Friedman SL: Fibrogenic cell reversion underlies fibrosis regression in liver. Proc Natl Acad Sci USA. 109:9230–9231. 2012.PubMed/NCBI View Article : Google Scholar

6 

Schuppan D: Structure of the extracellular matrix in normal and fibrotic liver: Collagens and glycoproteins. Semin Liver Dis. 10:1–10. 1990.PubMed/NCBI View Article : Google Scholar

7 

Herrera J, Henke CA and Bitterman PB: Extracellular matrix as a driver of progressive fibrosis. J Clin Invest. 128:45–53. 2018.PubMed/NCBI View Article : Google Scholar

8 

Anthony PP, Ishak KG, Nayak NC, Poulsen HE, Scheuer PJ and Sobin LH: The morphology of cirrhosis. Recommendations on definition, nomenclature, and classification by a working group sponsored by the World Health Organization. J Clin Pathol. 31:395–414. 1978.PubMed/NCBI View Article : Google Scholar

9 

Ginès P, Cárdenas A, Arroyo V and Rodés J: Management of cirrhosis and ascites. N Engl J Med. 350:1646–1654. 2004.PubMed/NCBI View Article : Google Scholar

10 

Zhou WC, Zhang QB and Qiao L: Pathogenesis of liver cirrhosis. World J Gastroenterol. 20:7312–7324. 2014.PubMed/NCBI View Article : Google Scholar

11 

Friedman SL, Roll FJ, Boyles J and Bissell DM: Hepatic lipocytes: The principal collagen-producing cells of normal rat liver. Proc Natl Acad Sci USA. 82:8681–8685. 1985.PubMed/NCBI View Article : Google Scholar

12 

Moreira RK: Hepatic stellate cells and liver fibrosis. Arch Pathol Lab Med. 131:1728–1734. 2007.PubMed/NCBI View Article : Google Scholar

13 

Yin C, Evason KJ, Asahina K and Stainier DY: Hepatic stellate cells in liver development, regeneration, and cancer. J Clin Invest. 123:1902–1910. 2013.PubMed/NCBI View Article : Google Scholar

14 

Bataller R and Brenner DA: Hepatic stellate cells as a target for the treatment of liver fibrosis. Semin Liver Dis. 21:437–451. 2001.PubMed/NCBI View Article : Google Scholar

15 

Tsuchida T and Friedman SL: Mechanisms of hepatic stellate cell activation. Nat Rev Gastroenterol Hepatol. 14:397–411. 2017.PubMed/NCBI View Article : Google Scholar

16 

Lanzoni G, Cardinale V and Carpino G: The hepatic, biliary, and pancreatic network of stem/progenitor cell niches in humans: A new reference frame for disease and regeneration. Hepatology. 64:277–286. 2016.PubMed/NCBI View Article : Google Scholar

17 

Kitade M, Kaji K and Yoshiji H: Relationship between hepatic progenitor cell-mediated liver regeneration and non-parenchymal cells. Hepatol Res. 46:1187–1193. 2016.PubMed/NCBI View Article : Google Scholar

18 

Boulter L, Govaere O, Bird TG, Radulescu S, Ramachandran P, Pellicoro A, Ridgway RA, Seo SS, Spee B, Van Rooijen N, et al: Macrophage-derived Wnt opposes Notch signaling to specify hepatic progenitor cell fate in chronic liver disease. Nat Med. 18:572–579. 2012.PubMed/NCBI View Article : Google Scholar

19 

Carpino G, Renzi A, Franchitto A, Cardinale V, Onori P, Reid L, Alvaro D and Gaudio E: Stem/progenitor cell niches involved in hepatic and biliary regeneration. Stem Cells Int. 2016(3658013)2016.PubMed/NCBI View Article : Google Scholar

20 

Grimaldi V, De Pascale MR, Zullo A, Soricelli A, Infante T, Mancini FP and Napoli C: Evidence of epigenetic tags in cardiac fibrosis. J Cardiol. 69:401–408. 2017.PubMed/NCBI View Article : Google Scholar

21 

Kopp F and Mendell JT: Functional classification and experimental dissection of long noncoding RNAs. Cell. 172:393–407. 2018.PubMed/NCBI View Article : Google Scholar

22 

Khorkova O, Hsiao J and Wahlestedt C: Basic biology and therapeutic implications of lncRNA. Adv Drug Deliv Rev. 87:15–24. 2015.PubMed/NCBI View Article : Google Scholar

23 

El Khodiry A, Afify M and El Tayebi HM: Behind the curtain of non-coding RNAs; long non-coding RNAs regulating hepatocarcinogenesis. World J Gastroenterol. 24:549–572. 2018.PubMed/NCBI View Article : Google Scholar

24 

Iyer MK, Niknafs YS, Malik R, Singhal U, Sahu A, Hosono Y, Barrette TR, Prensner JR, Evans JR, Zhao S, et al: The landscape of long noncoding RNAs in the human transcriptome. Nat Genet. 47:199–208. 2015.PubMed/NCBI View Article : Google Scholar

25 

Djebali S, Davis CA, Merkel A, Dobin A, Lassmann T, Mortazavi A, Tanzer A, Lagarde J, Lin W, Schlesinger F, et al: Landscape of transcription in human cells. Nature. 489:101–108. 2012.PubMed/NCBI View Article : Google Scholar

26 

Fatica A and Bozzoni I: Long non-coding RNAs: New players in cell differentiation and development. Nat Rev Genet. 15:7–21. 2014.PubMed/NCBI View Article : Google Scholar

27 

Ponting CP, Oliver PL and Reik W: Evolution and functions of long noncoding RNAs. Cell. 136:629–641. 2009.PubMed/NCBI View Article : Google Scholar

28 

Jiang X, Lei R and Ning Q: Circulating long noncoding RNAs as novel biomarkers of human diseases. Biomark Med. 10:757–769. 2016.PubMed/NCBI View Article : Google Scholar

29 

Hellerbrand C, Stefanovic B, Giordano F, Burchardt ER and Brenner DA: The role of TGFbeta1 in initiating hepatic stellate cell activation in vivo. J Hepatol. 30:77–87. 1999.PubMed/NCBI View Article : Google Scholar

30 

Inagaki Y and Okazaki I: Emerging insights into Transforming growth factor beta Smad signal in hepatic fibrogenesis. Gut. 56:284–292. 2007.PubMed/NCBI View Article : Google Scholar

31 

Dooley S and ten Dijke P: TGF-β in progression of liver disease. Cell Tissue Res. 347:245–256. 2012.PubMed/NCBI View Article : Google Scholar

32 

Fabregat I, Moreno-Càceres J, Sánchez A, Dooley S, Dewidar B, Giannelli G and Ten Dijke P: IT-LIVER Consortium. TGF-β signalling and liver disease. FEBS J. 283:2219–2232. 2016.PubMed/NCBI View Article : Google Scholar

33 

Breitkopf K, Godoy P, Ciuclan L, Singer MV and Dooley S: TGF-beta/Smad signaling in the injured liver. Z Gastroenterol. 44:57–66. 2006.PubMed/NCBI View Article : Google Scholar

34 

Friedman SL: Hepatic stellate cells: Protean, multifunctional, and enigmatic cells of the liver. Physiol Rev. 88:125–172. 2008.PubMed/NCBI View Article : Google Scholar

35 

Border WA and Noble NA: Evidence that TGF-beta should be a therapeutic target in diabetic nephropathy. Kidney Int. 54:1390–1391. 1998.PubMed/NCBI View Article : Google Scholar

36 

Fu N, Niu X, Wang Y, Du H, Wang B, Du J, Li Y, Wang R, Zhang Y, Zhao S, et al: Role of LncRNA-activated by transforming growth factor beta in the progression of hepatitis C virus-related liver fibrosis. Discov Med. 22:29–42. 2016.PubMed/NCBI

37 

Zhang K, Han X, Zhang Z, Zheng L, Hu Z, Yao Q, Cui H, Shu G, Si M, Li C, et al: The liver-enriched lnc-LFAR1 promotes liver fibrosis by activating TGFβ and Notch pathways. Nat Commun. 8(144)2017.PubMed/NCBI View Article : Google Scholar

38 

Zheng J, Mao Y, Dong P, Huang Z and Yu F: Long noncoding RNA HOTTIP mediates SRF expression through sponging miR-150 in hepatic stellate cells. J Cell Mol Med. 23:1572–1580. 2019.PubMed/NCBI View Article : Google Scholar

39 

Li Z, Wang J, Zeng Q, Hu C, Zhang J, Wang H, Yan J, Li H and Yu Z: Long noncoding RNA HOTTIP promotes mouse hepatic stellate cell activation via downregulating miR-148a. Cell Physiol Biochem. 51:2814–2828. 2018.PubMed/NCBI View Article : Google Scholar

40 

Jung KH, Zhang J, Zhou C, Shen H, Gagea M, Rodriguez-Aguayo C, Lopez-Berestein G, Sood AK and Beretta L: Differentiation therapy for hepatocellular carcinoma: Multifaceted effects of miR-148a on tumor growth and phenotype and liver fibrosis. Hepatology. 63:864–879. 2016.PubMed/NCBI View Article : Google Scholar

41 

Keniry A, Oxley D, Monnier P, Kyba M, Dandolo L, Smits G and Reik W: The H19 lincRNA is a developmental reservoir of miR-675 that suppresses growth and Igf1r. Nat Cell Biol. 14:659–665. 2012.PubMed/NCBI View Article : Google Scholar

42 

Chen X, Yamamoto M, Fujii K, Nagahama Y, Ooshio T, Xin B, Okada Y, Furukawa H and Nishikawa Y: Differential reactivation of fetal/neonatal genes in mouse liver tumors induced in cirrhotic and non-cirrhotic conditions. Cancer Sci. 106:972–981. 2015.PubMed/NCBI View Article : Google Scholar

43 

Zhang L, Zhou F, Drabsch Y, Gao R, Snaar-Jagalska BE, Mickanin C, Huang H, Sheppard KA, Porter JA, Lu CX and ten Dijke P: USP4 is regulated by AKT phosphorylation and directly deubiquitylates TGF-β type I receptor. Nat Cell Biol. 14:717–726. 2012.PubMed/NCBI View Article : Google Scholar

44 

Zhu J, Luo Z, Pan Y, Zheng W, Li W, Zhang Z, Xiong P, Xu D, Du M, Wang B, et al: H19/miR-148a/USP4 axis facilitates liver fibrosis by enhancing TGF-β signaling in both hepatic stellate cells and hepatocytes. J Cell Physiol. 234:9698–9710. 2019.PubMed/NCBI View Article : Google Scholar

45 

Zhou B, Yuan W and Li X: LncRNA Gm5091 alleviates alcoholic hepatic fibrosis by sponging miR-27b/23b/24 in mice. Cell Biol Int. 42:1330–1339. 2018.PubMed/NCBI View Article : Google Scholar

46 

Rogler CE, Matarlo JS, Kosmyna B, Fulop D and Rogler LE: Knockdown of miR-23, miR-27, and miR-24 alters fetal liver development and blocks fibrosis in mice. Gene Expr. 17:99–114. 2017.PubMed/NCBI View Article : Google Scholar

47 

Zhu D, Lyu L, Shen P, Wang J, Chen J, Sun X, Chen L, Zhang L, Zhou Q and Duan Y: rSjP40 protein promotes PPARγ expression in LX-2 cells through microRNA-27b. FASEB J. 32:4798–4803. 2018.PubMed/NCBI View Article : Google Scholar

48 

Clemson CM, Hutchinson JN, Sara SA, Ensminger AW, Fox AH, Chess A and Lawrence JB: An architectural role for a nuclear noncoding RNA: NEAT1 RNA is essential for the structure of paraspeckles. Mol Cell. 33:717–726. 2009.PubMed/NCBI View Article : Google Scholar

49 

Kong Y, Huang T, Zhang H, Zhang Q, Ren J, Guo X, Fan H and Liu L: The lncRNA NEAT1/miR-29b/Atg9a axis regulates IGFBPrP1-induced autophagy and activation of mouse hepatic stellate cells. Life Sci. 237(116902)2019.PubMed/NCBI View Article : Google Scholar

50 

Yu F, Jiang Z, Chen B, Dong P and Zheng J: NEAT1 accelerates the progression of liver fibrosis via regulation of microRNA-122 and Kruppel-like factor 6. J Mol Med (Berl). 95:1191–1202. 2017.PubMed/NCBI View Article : Google Scholar

51 

Zeng C, Wang YL, Xie C, Sang Y, Li TJ, Zhang M, Wang R, Zhang Q, Zheng L and Zhuang SM: Identification of a novel TGF-β-miR-122-fibronectin 1/serum response factor signaling cascade and its implication in hepatic fibrogenesis. Oncotarget. 6:12224–12233. 2015.PubMed/NCBI View Article : Google Scholar

52 

Kim Y, Ratziu V, Choi SG, Lalazar A, Theiss G, Dang Q, Kim SJ and Friedman SL: Transcriptional activation of transforming growth factor beta1 and its receptors by the Kruppel-like factor Zf9/core promoter-binding protein and Sp1. Potential mechanisms for autocrine fibrogenesis in response to injury. J Biol Chem. 273:33750–33758. 1998.PubMed/NCBI View Article : Google Scholar

53 

Tsai WC, Hsu SD, Hsu CS, Lai TC, Chen SJ, Shen R, Huang Y, Chen HC, Lee CH, Tsai TF, et al: MicroRNA-122 plays a critical role in liver homeostasis and hepatocarcinogenesis. J Clin Invest. 122:2884–2897. 2012.PubMed/NCBI View Article : Google Scholar

54 

Zhang K, Han Y, Hu Z, Zhang Z, Shao S, Yao Q, Zheng L, Wang J, Han X, Zhang Y, et al: SCARNA10, a nuclear-retained long non-coding RNA, promotes liver fibrosis and serves as a potential biomarker. Theranostics. 9:3622–3638. 2019.PubMed/NCBI View Article : Google Scholar

55 

Tu X, Zhang H, Zhang J, Zhao S, Zheng X, Zhang Z, Zhu J, Chen J, Dong L, Zang Y, et al: MicroRNA-101 suppresses liver fibrosis by targeting the TGFβ signalling pathway. J Pathol. 234:46–59. 2014.PubMed/NCBI View Article : Google Scholar

56 

Rapicavoli NA, Qu K, Zhang J, Mikhail M, Laberge RM and Chang HY: A mammalian pseudogene lncRNA at the interface of inflammation and anti-inflammatory therapeutics. Elife. 2(e00762)2013.PubMed/NCBI View Article : Google Scholar

57 

Nusslein-Volhard C and Wieschaus E: Mutations affecting segment number and polarity in Drosophila. Nature. 287:795–801. 1980.PubMed/NCBI View Article : Google Scholar

58 

Omenetti A, Choi S, Michelotti G and Diehl AM: Hedgehog signaling in the liver. J Hepatol. 54:366–373. 2011.PubMed/NCBI View Article : Google Scholar

59 

Machado MV and Diehl AM: Hedgehog signalling in liver pathophysiology. J Hepatol. 68:550–562. 2018.PubMed/NCBI View Article : Google Scholar

60 

Gorojankina T: Hedgehog signaling pathway: A novel model and molecular mechanisms of signal transduction. Cell Mol Life Sci. 73:1317–1332. 2016.PubMed/NCBI View Article : Google Scholar

61 

Sicklick JK, Li YX, Melhem A, Schmelzer E, Zdanowicz M, Huang J, Caballero M, Fair JH, Ludlow JW, McClelland RE, et al: Hedgehog signaling maintains resident hepatic progenitors throughout life. Am J Physiol Gastrointest Liver Physiol. 290:G859–G870. 2006.PubMed/NCBI View Article : Google Scholar

62 

Xie G, Choi SS, Syn WK, Michelotti GA, Swiderska M, Karaca G, Chan IS, Chen Y and Diehl AM: Hedgehog signalling regulates liver sinusoidal endothelial cell capillarisation. Gut. 62:299–309. 2013.PubMed/NCBI View Article : Google Scholar

63 

Syn WK, Agboola KM, Swiderska M, Michelotti GA, Liaskou E, Pang H, Xie G, Philips G, Chan IS, Karaca GF, et al: NKT-associated hedgehog and osteopontin drive fibrogenesis in non-alcoholic fatty liver disease. Gut. 61:1323–1329. 2012.PubMed/NCBI View Article : Google Scholar

64 

Yang L, Wang Y, Mao H, Fleig S, Omenetti A, Brown KD, Sicklick JK, Li YX and Diehl AM: Sonic hedgehog is an autocrine viability factor for myofibroblastic hepatic stellate cells. J Hepatol. 48:98–106. 2008.PubMed/NCBI View Article : Google Scholar

65 

Gao L, Zhang Z, Zhang P, Yu M and Yang T: Role of canonical Hedgehog signaling pathway in liver. Int J Biol Sci. 14:1636–1644. 2018.PubMed/NCBI View Article : Google Scholar

66 

Chen Y, Choi SS, Michelotti GA, Chan IS, Swiderska-Syn M, Karaca GF, Xie G, Moylan CA, Garibaldi F, Premont R, et al: Hedgehog controls hepatic stellate cell fate by regulating metabolism. Gastroenterology. 143:1319–1329.e11. 2012.PubMed/NCBI View Article : Google Scholar

67 

Briscoe J and Thérond PP: The mechanisms of Hedgehog signalling and its roles in development and disease. Nat Rev Mol Cell Biol. 14:416–429. 2013.PubMed/NCBI View Article : Google Scholar

68 

Syn WK, Jung Y, Omenetti A, Abdelmalek M, Guy CD, Yang L, Wang J, Witek RP, Fearing CM, Pereira TA, et al: Hedgehog-mediated epithelial-to-mesenchymal transition and fibrogenic repair in nonalcoholic fatty liver disease. Gastroenterology. 137:1478–1488.e8. 2009.PubMed/NCBI View Article : Google Scholar

69 

Sicklick JK, Li YX, Choi SS, Qi Y, Chen W, Bustamante M, Huang J, Zdanowicz M, Camp T, Torbenson MS, et al: Role for hedgehog signaling in hepatic stellate cell activation and viability. Lab Invest. 85:1368–1380. 2005.PubMed/NCBI View Article : Google Scholar

70 

Omenetti A, Yang L, Li YX, McCall SJ, Jung Y, Sicklick JK, Huang J, Choi S, Suzuki A and Diehl AM: Hedgehog-mediated mesenchymal-epithelial interactions modulate hepatic response to bile duct ligation. Lab Invest. 87:499–514. 2007.PubMed/NCBI View Article : Google Scholar

71 

Choi SS, Syn WK, Karaca GF, Omenetti A, Moylan CA, Witek RP, Agboola KM, Jung Y, Michelotti GA and Diehl AM: Leptin promotes the myofibroblastic phenotype in hepatic stellate cells by activating the hedgehog pathway. J Biol Chem. 285:36551–36560. 2010.PubMed/NCBI View Article : Google Scholar

72 

Choi SS, Omenetti A, Witek RP, Moylan CA, Syn WK, Jung Y, Yang L, Sudan DL, Sicklick JK, Michelotti GA, et al: Hedgehog pathway activation and epithelial-to-mesenchymal transitions during myofibroblastic transformation of rat hepatic cells in culture and cirrhosis. Am J Physiol Gastrointest Liver Physiol. 297:G1093–G1106. 2009.PubMed/NCBI View Article : Google Scholar

73 

Zheng J, Yu F, Dong P, Wu L, Zhang Y, Hu Y and Zheng L: Long non-coding RNA PVT1 activates hepatic stellate cells through competitively binding microRNA-152. Oncotarget. 7:62886–62897. 2016.PubMed/NCBI View Article : Google Scholar

74 

Yang JJ, Tao H, Huang C, Shi KH, Ma TT, Bian EB, Zhang L, Liu LP, Hu W, Lv XW and Li J: DNA methylation and MeCP2 regulation of PTCH1 expression during rats hepatic fibrosis. Cell Signal. 25:1202–1211. 2013.PubMed/NCBI View Article : Google Scholar

75 

Yu F, Lu Z, Chen B, Wu X, Dong P and Zheng J: Salvianolic acid B-induced microRNA-152 inhibits liver fibrosis by attenuating DNMT1-mediated Patched1 methylation. J Cell Mol Med. 19:2617–2632. 2015.PubMed/NCBI View Article : Google Scholar

76 

Yu F, Geng W, Dong P, Huang Z and Zheng J: LncRNA-MEG3 inhibits activation of hepatic stellate cells through SMO protein and miR-212. Cell Death Dis. 9(1014)2018.PubMed/NCBI View Article : Google Scholar

77 

Haertle L, Maierhofer A, Böck J, Lehnen H, Böttcher Y, Blüuher M, Schorsch M, Potabattula R, El Hajj N, Appenzeller S and Haaf T: Hypermethylation of the non-imprinted maternal MEG3 and paternal MEST alleles is highly variable among normal individuals. PLoS One. 12(e0184030)2017.PubMed/NCBI View Article : Google Scholar

78 

He Y, Wu YT, Huang C, Meng XM, Ma TT, Wu BM, Xu FY, Zhang L, Lv XW and Li J: Inhibitory effects of long noncoding RNA MEG3 on hepatic stellate cells activation and liver fibrogenesis. Biochim Biophys Acta. 1842:2204–2215. 2014.PubMed/NCBI View Article : Google Scholar

79 

He Y, Meng XM, Huang C, Wu BM, Zhang L, Lv XW and Li J: Long noncoding RNAs: Novel insights into hepatocelluar carcinoma. Cancer Lett. 344:20–27. 2014.PubMed/NCBI View Article : Google Scholar

80 

Logan CY and Nusse R: The Wnt signaling pathway in development and disease. Annu Rev Cell Dev Biol. 20:781–810. 2004.PubMed/NCBI View Article : Google Scholar

81 

Bejsovec A: Wnt signaling: An embarrassment of receptors. Curr Biol. 10:R919–R922. 2000.PubMed/NCBI View Article : Google Scholar

82 

Habas R and Dawid IB: Dishevelled and Wnt signaling: Is the nucleus the final frontier? J Biol. 4(2)2005.PubMed/NCBI View Article : Google Scholar

83 

Miller JR, Hocking AM, Brown JD and Moon RT: Mechanism and function of signal transduction by the Wnt/beta-catenin and Wnt/Ca2+ pathways. Oncogene. 18:7860–7872. 1999.PubMed/NCBI View Article : Google Scholar

84 

Kühl M, Sheldahl LC, Park M, Miller JR and Moon RT: The Wnt/Ca2+ pathway: A new vertebrate Wnt signaling pathway takes shape. Trends Genet. 16:279–283. 2000.PubMed/NCBI View Article : Google Scholar

85 

Veeman MT, Axelrod JD and Moon RT: A second canon. Functions and mechanisms of beta-catenin-independent Wnt signaling. Dev Cell. 5:367–377. 2003.PubMed/NCBI View Article : Google Scholar

86 

van Amerongen R, Mikels A and Nusse R: Alternative wnt signaling is initiated by distinct receptors. Sci Signal. 1(re9)2008.PubMed/NCBI View Article : Google Scholar

87 

Monga SP: beta-catenin signaling and roles in liver homeostasis, injury, and tumorigenesis. Gastroenterology. 148:1294–1310. 2015.PubMed/NCBI View Article : Google Scholar

88 

Rios-Esteves J and Resh MD: Stearoyl CoA desaturase is required to produce active, lipid-modified Wnt proteins. Cell Rep. 4:1072–1081. 2013.PubMed/NCBI View Article : Google Scholar

89 

Zhao C, Zhang M, Liu W, Wang C, Zhang Q and Li W: β-catenin knockdown inhibits pituitary adenoma cell proliferation and invasion via interfering with AKT and gelatinases expression. Int J Oncol. 46:1643–1650. 2015.PubMed/NCBI View Article : Google Scholar

90 

Xu W and Kimelman D: Mechanistic insights from structural studies of beta-catenin and its binding partners. J Cell Sci. 120:3337–3344. 2007.PubMed/NCBI View Article : Google Scholar

91 

Zhu Y, Tan J, Xie H, Wang J, Meng X and Wang R: HIF-1α regulates EMT via the Snail and beta-catenin pathways in paraquat poisoning-induced early pulmonary fibrosis. J Cell Mol Med. 20:688–697. 2016.PubMed/NCBI View Article : Google Scholar

92 

Thompson MD and Monga SP: WNT/beta-catenin signaling in liver health and disease. Hepatology. 45:1298–1305. 2007.PubMed/NCBI View Article : Google Scholar

93 

Miller JR: The Wnts. Genome Biol. 3(REVIEWS3001)2002.PubMed/NCBI View Article : Google Scholar

94 

Miao CG, Yang YY, He X, Huang C, Huang Y, Zhang L, Lv XW, Jin Y and Li J: Wnt signaling in liver fibrosis: Progress, challenges and potential directions. Biochimie. 95:2326–2335. 2013.PubMed/NCBI View Article : Google Scholar

95 

Ge WS, Wang YJ, Wu JX, Fan JG, Chen YW and Zhu L: β-catenin is overexpressed in hepatic fibrosis and blockage of Wnt/β-catenin signaling inhibits hepatic stellate cell activation. Mol Med Rep. 9:2145–2151. 2014.PubMed/NCBI View Article : Google Scholar

96 

Osawa Y, Oboki K, Imamura J, Kojika E, Hayashi Y, Hishima T, Saibara T, Shibasaki F, Kohara M and Kimura K: Inhibition of cyclic adenosine monophosphate (cAMP)-response element-binding protein (CREB)-binding protein (CBP)/β-catenin reduces liver fibrosis in mice. EBioMedicine. 2:1751–1758. 2015.PubMed/NCBI View Article : Google Scholar

97 

Kordes C, Sawitza I and Haussinger D: Canonical Wnt signaling maintains the quiescent stage of hepatic stellate cells. Biochem Biophys Res Commun. 367:116–123. 2008.PubMed/NCBI View Article : Google Scholar

98 

Yin X, Yi H, Wang L, Wu W, Wu X and Yu L: RSPOs facilitated HSC activation and promoted hepatic fibrogenesis. Oncotarget. 7:63767–63778. 2016.PubMed/NCBI View Article : Google Scholar

99 

Corbett L, Mann J and Mann DA: Non-canonical Wnt predominates in activated rat hepatic stellate cells, influencing HSC survival and paracrine stimulation of kupffer cells. PLoS One. 10(e0142794)2015.PubMed/NCBI View Article : Google Scholar

100 

Chatani N, Kamada Y, Kizu T, Ogura S, Furuta K, Egawa M, Hamano M, Ezaki H, Kiso S, Shimono A, et al: Secreted frizzled-related protein 5 (Sfrp5) decreases hepatic stellate cell activation and liver fibrosis. Liver Int. 35:2017–2026. 2015.PubMed/NCBI View Article : Google Scholar

101 

Shi SJ, Wang LJ, Yu B, Li YH, Jin Y and Bai XZ: LncRNA-ATB promotes trastuzumab resistance and invasion-metastasis cascade in breast cancer. Oncotarget. 6:11652–11663. 2015.PubMed/NCBI View Article : Google Scholar

102 

Li J, Li Z, Zheng W, Li X, Wang Z, Cui Y and Jiang X: LncRNA-ATB: An indispensable cancer-related long noncoding RNA. Cell Prolif. 50(e12381)2017.PubMed/NCBI View Article : Google Scholar

103 

Yuan JH, Yang F, Wang F, Ma JZ, Guo YJ, Tao QF, Liu F, Pan W, Wang TT, Zhou CC, et al: A long noncoding RNA activated by TGF-beta promotes the invasion-metastasis cascade in hepatocellular carcinoma. Cancer Cell. 25:666–681. 2014.PubMed/NCBI View Article : Google Scholar

104 

Liu J, Ruan B, You N, Huang Q, Liu W, Dang Z, Xu W, Zhou T, Ji R, Cao Y, et al: Downregulation of miR-200a induces EMT phenotypes and CSC-like signatures through targeting the β-catenin pathway in hepatic oval cells. PLoS One. 8(e79409)2013.PubMed/NCBI View Article : Google Scholar

105 

Su J, Zhang A, Shi Z, Ma F, Pu P, Wang T, Zhang J, Kang C and Zhang Q: MicroRNA-200a suppresses the Wnt/β-catenin signaling pathway by interacting with β-catenin. Int J Oncol. 40:1162–1170. 2012.PubMed/NCBI View Article : Google Scholar

106 

Fu N, Zhao SX, Kong LB, Du JH, Ren WG, Han F, Zhang QS, Li WC, Cui P, Wang RQ, et al: LncRNA-ATB/microRNA-200a/β-catenin regulatory axis involved in the progression of HCV-related hepatic fibrosis. Gene. 618:1–7. 2017.PubMed/NCBI View Article : Google Scholar

107 

Yu F, Zhou G, Huang K, Fan X, Li G, Chen B, Dong P and Zheng J: Serum lincRNA-p21 as a potential biomarker of liver fibrosis in chronic hepatitis B patients. J Viral Hepat. 24:580–588. 2017.PubMed/NCBI View Article : Google Scholar

108 

Yu F, Dong P, Mao Y, Zhao B, Huang Z and Zheng J: Loss of lncRNA-SNHG7 Promotes the Suppression of Hepatic Stellate Cell Activation via miR-378a-3p and DVL2. Mol Ther Nucleic Acids. 17:235–244. 2019.PubMed/NCBI View Article : Google Scholar

109 

Chen W, Zhao W, Yang A, Xu A, Wang H, Cong M, Liu T, Wang P and You H: Integrated analysis of microRNA and gene expression profiles reveals a functional regulatory module associated with liver fibrosis. Gene. 636:87–95. 2017.PubMed/NCBI View Article : Google Scholar

110 

Yao X, Liu C, Liu C, Xi W, Sun S and Gao Z: lncRNA SNHG7 sponges miR-425 to promote proliferation, migration, and invasion of hepatic carcinoma cells via Wnt/β-catenin/EMT signalling pathway. Cell Biochem Funct. 37:525–533. 2019.PubMed/NCBI View Article : Google Scholar

111 

Chakraborty JB and Mann DA: NF-kappaB signalling: Embracing complexity to achieve translation. J Hepatol. 52:285–291. 2010.PubMed/NCBI View Article : Google Scholar

112 

Ghosh S and Karin M: Missing pieces in the NF-kappaB puzzle. Cell. 109 (Suppl):S81–S96. 2002.PubMed/NCBI View Article : Google Scholar

113 

Sen R and Baltimore D: Multiple nuclear factors interact with the immunoglobulin enhancer sequences. Cell. 46:705–716. 1986.PubMed/NCBI View Article : Google Scholar

114 

Taniguchi K and Karin M: NF-κB, inflammation, immunity and cancer: Coming of age. Nat Rev Immunol. 18:309–324. 2018.PubMed/NCBI View Article : Google Scholar

115 

Gilmore TD: Introduction to NF-kappaB: Players, pathways, perspectives. Oncogene. 25:6680–6684. 2006.PubMed/NCBI View Article : Google Scholar

116 

Olefsky JM and Glass CK: Macrophages, inflammation, and insulin resistance. Annu Rev Physiol. 72:219–246. 2010.PubMed/NCBI View Article : Google Scholar

117 

Luedde T and Schwabe RF: NF-κB in the liver-linking injury, fibrosis and hepatocellular carcinoma. Nat Rev Gastroenterol Hepatol. 8:108–118. 2011.PubMed/NCBI View Article : Google Scholar

118 

Duffield JS, Forbes SJ, Constandinou CM, Clay S, Partolina M, Vuthoori S, Wu S, Lang R and Iredale JP: Selective depletion of macrophages reveals distinct, opposing roles during liver injury and repair. J Clin Invest. 115:56–65. 2005.PubMed/NCBI View Article : Google Scholar

119 

Pradere JP, Kluwe J, De Minicis S, Jiao JJ, Gwak GY, Dapito DH, Jang MK, Guenther ND, Mederacke I, Friedman R, et al: Hepatic macrophages but not dendritic cells contribute to liver fibrosis by promoting the survival of activated hepatic stellate cells in mice. Hepatology. 58:1461–1473. 2013.PubMed/NCBI View Article : Google Scholar

120 

Lv P, Luo HS, Zhou XP, Xiao YJ, Paul SC, Si XM and Zhou YH: Reversal effect of thalidomide on established hepatic cirrhosis in rats via inhibition of nuclear factor-kappaB/inhibitor of nuclear factor-kappaB pathway. Arch Med Res. 38:15–27. 2007.PubMed/NCBI View Article : Google Scholar

121 

Oakley F, Meso M, Iredale JP, Green K, Marek CJ, Zhou X, May MJ, Millward-Sadler H, Wright MC and Mann DA: Inhibition of inhibitor of kappaB kinases stimulates hepatic stellate cell apoptosis and accelerated recovery from rat liver fibrosis. Gastroenterology. 128:108–120. 2005.PubMed/NCBI View Article : Google Scholar

122 

Wright MC, Issa R, Smart DE, Trim N, Murray GI, Primrose JN, Arthur MJ, Iredale JP and Mann DA: Gliotoxin stimulates the apoptosis of human and rat hepatic stellate cells and enhances the resolution of liver fibrosis in rats. Gastroenterology. 121:685–698. 2001.PubMed/NCBI View Article : Google Scholar

123 

Seki E, De Minicis S, Osterreicher CH, Kluwe J, Osawa Y, Brenner DA and Schwabe RF: TLR4 enhances TGF-beta signaling and hepatic fibrosis. Nat Med. 13:1324–1332. 2007.PubMed/NCBI View Article : Google Scholar

124 

Sunami Y, Leithauser F, Gul S, Fiedler K, Guldiken N, Espenlaub S, Holzmann KH, Hipp N, Sindrilaru A, Luedde T, et al: Hepatic activation of IKK/NFκB signaling induces liver fibrosis via macrophage-mediated chronic inflammation. Hepatology. 56:1117–1128. 2012.PubMed/NCBI View Article : Google Scholar

125 

Shen H, Sheng L, Chen Z, Jiang L, Su H, Yin L, Omary MB and Rui L: Mouse hepatocyte overexpression of NF-κB-inducing kinase (NIK) triggers fatal macrophage-dependent liver injury and fibrosis. Hepatology. 60:2065–2076. 2014.PubMed/NCBI View Article : Google Scholar

126 

Son G, Iimuro Y, Seki E, Hirano T, Kaneda Y and Fujimoto J: Selective inactivation of NF-kappaB in the liver using NF-kappaB decoy suppresses CCl4-induced liver injury and fibrosis. Am J Physiol Gastrointest Liver Physiol. 293:G631–G639. 2007.PubMed/NCBI View Article : Google Scholar

127 

Zhang H, Li H, Ge A, Guo E, Liu S and Zhang L: Long non-coding RNA TUG1 inhibits apoptosis and inflammatory response in LPS-treated H9c2 cells by down-regulation of miR-29b. Biomed Pharmacother. 101:663–669. 2018.PubMed/NCBI View Article : Google Scholar

128 

Roderburg C, Urban GW, Bettermann K, Vucur M, Zimmermann H, Schmidt S, Janssen J, Koppe C, Knolle P, Castoldi M, et al: Micro-RNA profiling reveals a role for miR-29 in human and murine liver fibrosis. Hepatology. 53:209–218. 2011.PubMed/NCBI View Article : Google Scholar

129 

Sekiya Y, Ogawa T, Yoshizato K, Ikeda K and Kawada N: Suppression of hepatic stellate cell activation by microRNA-29b. Biochem Biophys Res Commun. 412:74–79. 2011.PubMed/NCBI View Article : Google Scholar

130 

Ogawa T, Iizuka M, Sekiya Y, Yoshizato K, Ikeda K and Kawada N: Suppression of type I collagen production by microRNA-29b in cultured human stellate cells. Biochem Biophys Res Commun. 391:316–321. 2010.PubMed/NCBI View Article : Google Scholar

131 

Xing TJ, Jiang DF, Huang JX and Xu ZL: Expression and clinical significance of miR-122 and miR-29 in hepatitis B virus-related liver disease. Genet Mol Res. 13:7912–7918. 2014.PubMed/NCBI View Article : Google Scholar

132 

Han X, Hong Y and Zhang K: TUG1 is involved in liver fibrosis and activation of HSCs by regulating miR-29b. Biochem Biophys Res Commun. 503:1394–1400. 2018.PubMed/NCBI View Article : Google Scholar

133 

Geisler F and Strazzabosco M: Emerging roles of Notch signaling in liver disease. Hepatology. 61:382–392. 2015.PubMed/NCBI View Article : Google Scholar

134 

Morell CM and Strazzabosco M: Notch signaling and new therapeutic options in liver disease. J Hepatol. 60:885–890. 2014.PubMed/NCBI View Article : Google Scholar

135 

Siebel C and Lendahl U: Notch signaling in development, tissue homeostasis, and disease. Physiol Rev. 97:1235–1294. 2017.PubMed/NCBI View Article : Google Scholar

136 

Wakabayashi N, Chartoumpekis DV and Kensler TW: Crosstalk between Nrf2 and Notch signaling. Free Radic Biol Med. 88:158–167. 2015.PubMed/NCBI View Article : Google Scholar

137 

Ni MM, Wang YR, Wu WW, Xia CC, Zhang YH, Xu J, Xu T and Li J: Novel Insights on Notch signaling pathways in liver fibrosis. Eur J Pharmacol. 826:66–74. 2018.PubMed/NCBI View Article : Google Scholar

138 

Kimball AS, Joshi AD, Boniakowski AE, Schaller M, Chung J, Allen R, Bermick J, Carson WF IV, Henke PK, Maillard I, et al: Notch regulates macrophage-mediated inflammation in diabetic wound healing. Front Immunol. 8(635)2017.PubMed/NCBI View Article : Google Scholar

139 

Wang T, Xiang Z, Wang Y, Li X, Fang C, Song S, Li C, Yu H, Wang H, Yan L, et al: (-)-Epigallocatechin gallate targets notch to attenuate the inflammatory response in the immediate early stage in human macrophages. Front Immunol. 8(433)2017.PubMed/NCBI View Article : Google Scholar

140 

Xie G, Karaca G, Swiderska-Syn M, Michelotti GA, Kruger L, Chen Y, Premont RT, Choi SS and Diehl AM: Cross-talk between Notch and Hedgehog regulates hepatic stellate cell fate in mice. Hepatology. 58:1801–1813. 2013.PubMed/NCBI View Article : Google Scholar

141 

Romeo S: Notch and nonalcoholic fatty liver and fibrosis. N Engl J Med. 380:681–683. 2019.PubMed/NCBI View Article : Google Scholar

142 

Zhu C, Kim K, Wang X, Bartolome A, Salomao M, Dongiovanni P, Meroni M, Graham MJ, Yates KP, Diehl AM, et al: Hepatocyte Notch activation induces liver fibrosis in nonalcoholic steatohepatitis. Sci Transl Med. 10(eaat0344)2018.PubMed/NCBI View Article : Google Scholar

143 

Iso T, Kedes L and Hamamori Y: HES and HERP families: Multiple effectors of the Notch signaling pathway. J Cell Physiol. 194:237–255. 2003.PubMed/NCBI View Article : Google Scholar

144 

Kageyama R, Ohtsuka T, Hatakeyama J and Ohsawa R: Roles of bHLH genes in neural stem cell differentiation. Exp Cell Res. 306:343–348. 2005.PubMed/NCBI View Article : Google Scholar

145 

Yu F, Chen B, Dong P and Zheng J: HOTAIR epigenetically modulates PTEN expression via MicroRNA-29b: A novel mechanism in regulation of liver fibrosis. Mol Ther. 25:205–217. 2017.PubMed/NCBI View Article : Google Scholar

146 

Dong Z, Li S, Wang X, Si L, Ma R, Bao L and Bo A: lncRNA GAS5 restrains CCl4-induced hepatic fibrosis by targeting miR-23a through the PTEN/PI3K/Akt signaling pathway. Am J Physiol Gastrointest Liver Physiol. 316:G539–G550. 2019.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

April-2021
Volume 21 Issue 4

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wu Z, Huang S, Zheng X, Gu S, Xu Q, Gong Y, Zhang J, Fu B and Tang L: Regulatory long non‑coding RNAs of hepatic stellate cells in liver fibrosis (Review). Exp Ther Med 21: 351, 2021
APA
Wu, Z., Huang, S., Zheng, X., Gu, S., Xu, Q., Gong, Y. ... Tang, L. (2021). Regulatory long non‑coding RNAs of hepatic stellate cells in liver fibrosis (Review). Experimental and Therapeutic Medicine, 21, 351. https://doi.org/10.3892/etm.2021.9782
MLA
Wu, Z., Huang, S., Zheng, X., Gu, S., Xu, Q., Gong, Y., Zhang, J., Fu, B., Tang, L."Regulatory long non‑coding RNAs of hepatic stellate cells in liver fibrosis (Review)". Experimental and Therapeutic Medicine 21.4 (2021): 351.
Chicago
Wu, Z., Huang, S., Zheng, X., Gu, S., Xu, Q., Gong, Y., Zhang, J., Fu, B., Tang, L."Regulatory long non‑coding RNAs of hepatic stellate cells in liver fibrosis (Review)". Experimental and Therapeutic Medicine 21, no. 4 (2021): 351. https://doi.org/10.3892/etm.2021.9782