Abstract
Liver fibrosis is characterized by the transdifferentiation of hepatic stellate cells (HSCs) to myofibroblasts and poor response to treatment. This can be attributed to the myofibroblast-specific resistance to phenotype reversal. In this study, we complemented miR-16 into miR-16-deficient myofibroblasts and analyzed the global role of miR-16 using transcriptome profiling and generating a pathway-based action model underlying transcriptomic regulation. Phenotypic analysis of myofibroblasts and fibrogenic characterization were used to understand the effect of miR-16 on phenotypic remodeling of myofibroblasts. miR-16 expression altered the transcriptome of myofibroblasts to resemble that of HSCs. Simultaneous targeting of Smad2 and Wnt3a, etc. by miR-16 integrated signaling pathways of TGF-β and Wnt, etc., which underlay the comprehensive regulation of transcriptome. The synergistic effect of miR-16 on the signaling pathways abolished the phenotypic characteristics of myofibroblasts, including collagen production and inhibition of adipogenesis. In vivo, myofibroblast-specific expression of miR-16 not only eliminated mesenchymal cells with myofibroblast characteristics but also restored the phenotype of HSCs in perisinusoidal space. This phenotypic remodeling resolved liver fibrosis induced by chronic wound healing. Therefore, miR-16 may integrate signaling pathways crucial for the fate determination of myofibroblasts. Its global effect induces the reversal of HSC-to-myofibroblast transdifferentiation and, subsequently, the resolution of fibrogenesis. Taken together, these findings highlight the potential of miR-16 as a promising therapeutic target for liver fibrosis.
Subject terms: Liver diseases, Liver fibrosis
Introduction
Liver fibrosis, a serious health problem worldwide, is induced by chronic liver injuries (hepatitis, alcoholism, cholestasis, etc.), which often lead to the development of hepatocellular carcinoma1. Liver fibrosis is characterized by the transdifferentiation of hepatic stellate cells (HSCs), a kind of adipogenic cells, toward myofibroblasts2. Multiple signaling pathways act in concert to enable this phenotypic transition2,3. Thus, such a complex network makes it difficult to target a single biomolecule, reverse the HSC-to-myofibroblast transdifferentiation, and ensure good response to treatment.
In contrast to most agents that act through single target, miRNA demonstrates a unique pattern, mainly multiple targeting, of gene regulation4. This pleiotropic effect highlights the importance of miRNAs in the phenotypic determination of various types of cells5–9. Among phenotype-regulating miRNAs, miR-16 is the most extensively explored one ever since its identification in 200510,11, and serves as internal control of miRNA analysis for an universal and relative stable expression12. miR-16 undergoes loss of expression during the transdifferentiation of HSCs13. Its deficiency correlates to the myofibroblast-specific phenotype and apoptosis resistance, via Bcl-2-targeted activation of the mitochondrial apoptosis pathway13. Therefore, miR-16 is highlighted to be a potential in the phenotype controlling of myofibroblasts. However, whether miR-16 synergistically regulates multiple targets in a disease- or phenotype-specific manner and its association with the global regulation of mRNAs remain to be understood. The numerous targets of miR-16 had lent increasing ambiguity to its global effect, thereby making it difficult to understand the role of miR-16 in the resolution of fibrosis.
Muscle-specific miR-145 has been recently reported to function during the conversion of adult fibroblasts into smooth muscle cells with a contractile phenotype6,7. miR-206 (also muscle-specific) blocks the maturation of osteoblasts8 and growth of rhabdomyosarcoma by targeting myogenic differentiation14. Neurogenetic miRNAs, miR-124a and miR-125b, also help in the formation of mature neurons from the neuroblastoma line SH-SY5Y and neural progenitor ReNcell VM9. Thus, miRNAs function in a synergistic manner. miR-34c overexpression in HeLa cells upregulates germ cell-specific genes15. miR-1 alters the transcriptome of HeLa cells to resemble that of muscle cells16. Depletion of miR-23b during hepatocyte differentiation promotes the expression of bile duct-specific genes17. Therefore, transcriptome regulation may underlie the global action of miRNAs. Despite affecting multiple genes, miRNAs show limited base-pairing to the miRNA-regulated genes18–21. miRNAs possess preference for signaling pathway targets that highlights their importance as therapeutic targets22–24. Accordingly, we hypothesized that miRNAs exert a global cell-specific effect on the transcriptome and phenotype by integrated regulation of signaling pathways.
To test the synergistic actions of miR-16, we restored its expression in myofibroblasts and evaluated its impact on the cellular transcriptome. We used bioinformatic modeling to identify the central signaling pathways targeted to regulate the transcriptome. Wnt and transforming growth factor (TGF)-β signaling involved in adipogenesis and collagen production, respectively, revealed the role of miR-16 in myofibroblast transdifferentiation by integration of signaling pathways. The global effect of miR-16 on liver fibrosis was assessed on the basis of phenotype remodeling of myofibroblast.
Materials and methods
Isolation and identification of HSCs and myofibroblasts
HSCs were isolated from normal rats using in situ perfusion and density-gradient centrifugation25. Nevertheless, myofibroblasts were separated from rats with CCl4-induced liver fibrosis (Supplementary Fig. S1) by liver section and type IV collagenase/Pronase E/DNAse digestion. Freshly isolated myofibroblasts were purified using sequential centrifugation through 13% and 11% Nycodenz26. After 24 h culture for homogeneity, the viability (over 95%) and purity (up to 90%) of both HSCs and myofibroblasts were verified (Supplementary Fig. S2). Cell morphology and phenotype was assayed using oil red O staining and immunofluorescence for biomarkers (α-SMA and desmin) (Supplementary Fig. S2)13,25,26.
Restoration of miR-16 levels in myofibroblasts
Rat HSCs and myofibroblasts were divided into seven groups at random, namely HSC, myofibroblast, pLV-GFP-treated myofibroblast, pLV-miR-16-treated myofibroblast, miR-16 inhibitor-treated myofibroblast, Smad2 small interfering RNA (siRNA)-treated myofibroblast, and Wnt3a siRNA-treated myofibroblast (n = 9 samples/group). The pLV-miR-16-treated and pLV-GFP-treated groups were infected with pLV-miR-16 (containing pre-rno-miR-16) and pLV-GFP (containing pre-rno-miR-16 with 5′-GGGGGG-3′ instead of the seed sequence), respectively, at 1.0 × 108 transduction unit (TU)/ml and multiplicity of infection of 30. The group of miR-16 inhibitor-treated myofibroblast was administrated by both 20 µM miR-16 inhibitor (GenePharm, Shanghai, China) and pLV-miR-16 as mentioned above. Whereas Smad2 siRNA (GenePharm, Shanghai, China), and Wnt3a siRNA (GenePharm, Shanghai, China) were transfected into groups of Smad2 siRNA-treated myofibroblast and Wnt3a siRNA-treated myofibroblast, respectively, according to the manufacturer’s instructions.
Total RNA was extracted from cells 24 h after isolation (HSC group and myofibroblast group of rats, HSC, and myofibroblast of human) or 48 h post transduction (pLV-miR-16-treated myofibroblast group, pLV-GFP-treated myofibroblast group). Thereafter, we performed stem-loop quantitative reverse transcription-PCR (RT-QPCR) for miR-16 and miR-15b of the miR-16 cluster using the TaqMan MicroRNA Assay (Applied Biosystems, Foster City, CA, USA). miRNA expression was normalized to U87 snRNA levels.
Microarray analysis and bioinformatic modeling of miR-16
Transcriptome profiling was performed using Affymetrix rat 230 2.0 arrays (Affymetrix, Santa Clara, CA, USA) 24 h after cell isolation (groups of HSC and myofibroblast) or 12–48 h after lentiviral transfection (groups of pLV-miR-16-treated and pLV-GFP-treated myofibroblast).
The predicted target gene sets of miR-16 were obtained from databases of miRBase and TargetScan 5.1, and intersected with miR-16-regulated gene set. These gene sets, after being supplemented with proven targets, were mapped to DAVID database to generate sets of signaling pathways27. The intersection between three sets of signaling pathways have been presented. Furthermore, signaling pathways relevant to miR-16 were filtered from the intersection upon enrichment against the background. Their roles and relationships were measured by biological-process-based categorization and generation of pathway–pathway interaction network, according to the annotations of the Kyoto Encyclopedia of Genes and Genomes (KEGG) and directed graph theory topology. Mediating the effect of most other signaling pathways (>3), 20% signaling pathways were classified as the critical nodes of pathway–pathway interaction network and the key executors of miR-16’s actions.
Effect of miR-16 on targets within signaling pathways
We performed luciferase assays to determine the targeted effect of miR-16 on SMAD2 and Wnt3a. Transcription of the targets (genes involved in signaling pathways and their downstream genes) of miR-16 were subsequently detected in seven groups of HSCs and myofibroblasts using RT-QPCR (Supplementary Table S1). The in vivo and in vitro expression of the targets were analyzed using immunofluorescence and quantified of western blotting.
Functional analysis of myofibroblasts upon miR-16 restoration
Enzyme-linked immunosorbent assays (Cusabio, College Park, MD, USA) were used to determine the levels of collagen type I and III in the culture supernatants of seven groups of HSCs and myofibroblasts. Cell cycle progression and proliferation were assayed using propidium iodide staining and the Cell Counting Kit 8 (Dojindo, Kumamoto, Japan), respectively, after serum starvation for 24 h.
Moreover, the adipogenic activity of the seven groups of HSCs and myofibroblasts were assayed based on the expression of adipogenic transcription factors (TFs; PPARγ, C/EBPα, and RXRα) and normalized using β-catenin levels.
Phenotypic characterization of myofibroblasts after targeted miR-16 normalization
Rats were randomized into the normal control group (n = 6), fibrosis model group (n = 5), pLV-miR-16-treated group (n = 5), and pLV-GFP-treated group (n = 5). After the 40% CCl4 administration (0.3 ml/kg) for 4 weeks, pLV-miR-16 (pLV-miR-16-treated group, 1.0 × 108 TU/week) with α-SMA promoter28 or pLV-GFP (pLV-GFP-treated group, 1.2 × 108 TU/week) was delivered, respectively, in vivo by superior mesentery vein catheterization for another 4 weeks29. The liver-targeted lentiviral transfection, with or without miR-16, was assessed by monitoring the green fluorescent protein (GFP) signal.
At the end of 8 weeks, the presence of dual-labeled Wnt3a/α-SMA and Smad2/α-SMA mesenchymal cells were monitored in liver sections by immunofluorescence. Transmission electron microscopy was used to identify ultrastructures specific to HSCs and myofibroblasts. Apoptosis and proliferation were assayed using terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) and immunohistochemical labeling of proliferating cell nuclear antigen (PCNA), respectively.
Global effect of miR-16 on liver fibrosis
Hepatic histopathology of normal control, fibrosis model, pLV-miR-16-treated, and pLV-GFP-treated groups was assessed by hematoxylin and eosin staining and Sirius red staining. Collagen deposition was assayed using immunofluorescence and quantified by western blotting for collagen type I and III. Fibrosis staging was independently determined, in accordance with the Ishak staging system, by two pathologists who were not aware of the experiments. The protocols in study were approved by the Ethical Committee at Xinhua Hospital. Rats received humane care in accordance with the US Public Health Service Policy on Humane Care and Use of Laboratory Animals.
Statistical analysis
Data have been represented as mean ± SD. Independent t-test and analysis of variance were applied for pair comparisons and multiple comparisons, respectively. A χ2-test was done with quadruple tabular form. Sample randomness was determined using the equal probability sampling method (SPSS 15.0). Differences with P < 0.05 were considered statistically significant.
Results
Restoration of miR-16 shaped the transcriptome of myofibroblast towards HSC-like one
As compared to that in HSCs, we observed significant downregulation of miR-16 in the myofibroblasts (Supplementary Fig. S3). Rodent myofibroblasts were then transduced with pLV-miR-16 at an efficiency of over 90% (Supplementary Figs. S4 and S5) after their isolation, 24 h cultural homogeneity, and identification. Although few changes in the transcriptome were observed at the 12 and 24 h time points, the up- and downregulated transcripts reached 1162 (3.74%) and 920 (2.96%), respectively, at 48 h with the restoration of miR-16 level (Supplementary Fig. S6). miR-15b levels, another member of the miR-16 cluster, remained constant during the same period (Supplementary Fig. S6).
After validating the alterations in the transcriptome (Supplementary Fig. S7), the filtered genes were categorized by hierarchical clustering (Fig. 1a) and function (Supplementary Table S2). (1) Adipogenic genes, which characterizes HSCs, were increased after the administration of miR-16. (2) miR-16-regulated cytokines, collagens, and matrix metallopeptidases showed a rebalancing of extracellular matrix (ECM) production and zymohydrolysis, thereby inactivating the myofibroblasts and facilitating the regression of hepatic fibrosis. (3) Genes associated with cell cycle arrest in myofibroblasts were induced upon restoration of miR-16 expression, including the upregulation of cell cycle inhibitors and downregulation of cell cycle inducers. (4) A significant population of the upregulated transcripts participated in mitochondrial apoptosis; this could result in reduced apoptosis in myofibroblasts. In summary, the induction of miR-16 expression reversed the mRNA profile of myofibroblasts to that of HSCs.
Bioinformatic modeling identified the targeted effect of miR-16 on signaling pathways
Using the miRanda, TargetScan algorithm, and transcriptomic filtration, three sets of gene targets (miRanda-predicted targets, TargetScan-predicted targets, and transcriptome-filtered targets) were generated, which corresponded to the transcripts predicted or tested to be regulated by miR-16. The intersection between these three sets, however, comprised a limited number of genes (Fig. 1b). Notably, the three sets of signaling pathways (miRanda-predicted pathways, TargetScan-predicted pathways, and transcriptome-filtered pathways), predicted or validated to be regulated by miR-16, significantly overlapped with each other (Fig. 1c, P < 0.001).
Using Gene Ontology, we functionally annotated the miR-16-regulated genes (Supplementary Table S3). This included a major proportion of intracellular signaling cascades (7.04%, P = 0.001). In addition, 32.3% of the miR-16-regulated genes were annotated to be members of signaling pathways, significantly higher than the percentage of background (P < 0.001) (Supplementary Fig. S8). The integrated effect of a specific set of signaling pathways, therefore, seems to underlie the impact of miR-16 on the transcriptome.
Organization of the signaling pathways into functional categories revealed the synergistic effect of 43 intersected signaling pathways on the basis of KEGG annotation. Proportionally, Cancers, Signal Transduction, Cell Growth and Death, etc. were the most represented categories (Fig. 1d). Among these intersected pathways, eight signaling pathways were the most crucial nodes: Cell cycle, Apoptosis, TGF-β signaling pathway, Wnt signaling pathway, p53 signaling pathway, Focal adhesion, Ubiquitin-mediated Proteolysis, and ErbB signaling pathway, as they served as the nodes of the pathway–pathway interaction network (Fig. 1e).
miR-16 inhibited the fibrotic and proliferative properties of myofibroblasts by targeting TGF-β signaling pathway
Dual luciferase assays showed that miR-16 targeted SMAD2, a key member of the TGF-β signaling pathway (Fig. 2a, b). Protein and mRNA levels of SMAD2 decreased with the expression of miR-16 (Fig. 2c, d and Supplementary Fig. S9A). The mRNA levels of genes downstream to the TGF-β signaling pathway (collagen type I and III) also decreased (Fig. 2e). miR-16 reduced the concentrations of collagen type I and III in the culture supernatant, indicating net changes in ECM synthesis and secretion (Fig. 2f). Inactivation of TGF-β signaling pathway also induced the G0/G1 block and reduced G2/M proportion (Fig. 2g) of myofibroblasts, resulting in cell cycle arrest and inhibition of proliferation (Fig. 2h). Dramatically, the effects of miR-16 on ECM production and myofibroblasts proliferation were recapitulated by SMAD2-specific RNA interference and abrogated by miR-16 inhibitor (Fig. 2c–h and Supplementary Fig. S9A).
miR-16 restored adipogenic characteristics of myofibroblasts by targeting Wnt signaling pathway
Wnt3a, a component of Wnt signaling, was another target of miR-16 (Fig. 3a, b). Wnt3a was transcriptionally and translationally downregulated in pLV-miR-16-treated myofibroblasts (Fig. 3c, d and Supplementary Fig. S9B). This resulted in the reduced accumulation of β-catenin, thereby promoting the mesenchymal phenotype in myofibroblasts (Fig. 3e, f and Supplementary Fig. S10). On the contrary, an elevated expression of adipogenic TFs (C/EBPα, PPARγ, and RXRα) took place simultaneously with statistical significance (Fig. 3e, f and Supplementary Fig. S10). Moreover, depletion of Wnt3a in the myofibroblasts regulated the expression of mesenchymal markers and adipogenic TFs, mimicking those in miR-16-restored myofibroblasts (Fig. 3c–f and Supplementary Figs. S9B and S10). The miR-16 inhibitor reversed the changes in expression induced by miR-16 (Fig. 3c–f and Supplementary Figs. S9B and S10).
miR-16 induced phenotype reversal from myofibroblast to HSC-like cell by integrated effects on signaling pathways
miR-16 was downregulated in myofibroblasts as compared to that in HSCs derived from rats and patients with liver fibrosis (Fig. 4a, d). We also observed an increase in the mRNA and protein levels of Smad2 and Wnt3a (Figs. 4b, c, e, f and Supplementary Figs. S11 and S12). In contrast, myofibroblast-targeted in vivo miR-16 restoration (Supplementary Fig. S13), performing from week 5 to 8, mitigated the miR-16 lacking (fibrosis model group vs. pLV-miR-16-treated group, P < 0.01) and the overexpression of its targets (Fig. 4d–f and Supplementary Fig. S12).
The targeting effect of miR-16 on Smad2 and Wnt3a reduced mesenchymal cells dual labeling for Smad2/α-SMA and Wnt3a/α-SMA (Fig. 4g and Supplementary Fig. S14). Ultrastructural findings, especially the existence of lipid droplets instead of the deposition of collagen filament, and regaining the spherical-like instead of stretched morphology, indicated that miR-16 shaped myofibroblasts towards the phenotype of HSCs (Fig. 4h). Despite the chemotactic stimuli, miR-16-induced HSC-like cells were localized to the sinusoidal space without aggregation at the site of the injury.
Analysis of apoptosis and proliferation reflected additional effects of miR-16 on phenotypic modulation. By TUNEL, an augment of positive signals enriching these HSC-like cells were shown in the pLV-miR-16-treated group rather than fibrosis model group and pLV-GFP-treated group (Fig. 4i, j). However, the pLV-miR-16-treated group exhibited the reduced level of proliferative marker (PCNA) in comparison to other two groups (Fig. 4k, l).
miR-16-mediated myofibroblast remodeling resolved liver fibrosis
Figure 5 shows the miR-16-mediated phenotype remodeling of myofibroblasts. This was associated with the downregulation of collagen type I and III (Fig. 6a–d and Supplementary Figs. S15 and S16) and dissolution of fibrous cords (Fig. 6e, f). Furthermore, the global impact of miR-16 lead to a decrease in the fibrosis stage (fibrosis model group vs. pLV-miR-16-treated group: 4.93 ± 0.58 vs. 2.11 ± 0.57, P < 0.01; pLV-GFP-treated group vs. pLV-miR-16-treated group: 4.53 ± 0.73 vs. 2.11 ± 0.57, P < 0.01) (Fig. 6g). However, other markers of hepatic injury (hepatosteatosis, inflammation, etc.) were not affected by the miR-16 expression.
Discussion
miR-16 has been extensively studied as a regulator of cell phenotype13,30,31. Its deficiency demonstrates close association with HSC-myofibroblast transdifferentiation13. However, the global effect of miR-16 on cellular morphology and function remains to be understood. In this study, we observed the differential expression of multiple phenotype-determining genes upon the restoration of miR-16 expression at 48 h in myofibroblasts. HSC-specific adipogenic genes, including RXRα32–34, PPARγ32–35, CEBPα32,36,37, CEBPθ2,32, Fabp62,32, Fabp72,32, and Rbp238,39, etc., were upregulated with the expression of miR-16. miR-16 also downregulated major fibrosis-inducing cytokines and related genes (TGFβ240, LTBP441, etc.) and collagens (Col1a142, Col3a142, and Col4a543, etc.), whereas it upregulated fibrolysis-dependent matrix metallopeptidases (MMP-244, etc.). Furthermore, the decreased expression of CCND145, CCNY46, Cdc2047, Cdc4248, Cdc42ep149,50, and Cdc42ep249, together with the elevated transcription of CDKN1A51, each holding a critical place in cell cycle, reflected the inhibition of proliferation. Genes related to mitochondrial apoptosis, including Bcl2l1352, Casp353, and Casp954, etc., were overexpressed with increasing expression of miR-16. Attenuation of apoptosis resistance, the biological property of myofibroblasts, was suggested. miR-16 expression reversed the transcriptional profile of myofibroblasts to that of HSCs. The stable expression of miR-15b, another important member of miR-16 cluster, during 0–48 h convinced the miR-16 property of transcriptional regulation.
Signaling pathways regulated by miR-16 were acquired by the transcriptomic findings and two predicted target sets using DAVID-based data mapping. In contrast to the limited intersection between miR-16-regulated genes and predicted target genes, different sets of signaling pathways shared a large proportion of components. Therefore, synergistic regulation of a certain set of signaling pathways underlay the transcriptomic regulation of miR-16. Functional categorization using KEGG further outlined the global action of miR-16 through the intersected set of signaling pathways. By percentage, Cancers (27.91%), Signal Transduction (11.63%), Cell Growth and Death (9.30%), immune system (6.98%), Cell Communication (4.65%), and Lipid Metabolism (4.65%), etc. were classified to be the most significant functions of miR-16-regulated signaling pathways. This is in accordance with the function of miR-16 as described in other studies13,21,55.
We generated a pathway–pathway interaction network to identify the central signaling pathways that mediated the global effect of miR-16. The signaling pathways most regulated included the Wnt signaling pathway, TGF-β signaling pathway, p53 signaling pathway, ErbB signaling pathway, Apoptosis, Cell cycle, Focal adhesion, and Ubiquitin-mediated proteolysis. Among these, the signaling pathways related to TGF-β56, Wnt57, Apoptosis13, Cell cycle58, p5359, and Focal adhesion60 are involved in the transdifferentiation of HSCs to myofibroblasts. Thus, these pathways were proposed as the key and cooperative mediators for miR-16’s action on myofibroblasts.
To verify the established action model for miR-16, the key signaling pathways and predicted target genes were integrated and functionally analyzed. SMAD2 was a direct target of miR-16. There was a decrease in the mRNA and protein levels of SMAD2 with the increase in miR-16 levels, suggesting the negative effect of miR-16 on TGF-β signaling in myofibroblasts. This resulted in the inhibited expression and secretion of collagen types I and III. Moreover, miR-16-mediated suppression of TGF-β signaling pathway resulted in cell cycle arrest and inhibition of proliferation in myofibroblasts61. miR-16 also regulated Wnt signaling in these cells: Wnt3a, the activator of Wnt signaling pathway and anti-adipogenic morphogen62, was complementarily downregulated by miR-16. Subsequently, the expression of adipogenic TFs in HSC-like cells, which were epigenetically repressed in myofibroblasts in a β-catenin-dependent manner, was restored in the absence of Wnt-induced accumulation of β-catenin. Abolishment of apoptosis resistance and active proliferation reflected the other aspects of miR-16’s role, yet miR-16-based phenotypical remodeling underlies the outcome of most part of myofibroblast population. These findings with target-knockdown-based recapitulative and miR-16-inhibitor-based antagonistic proofs provided an explanation for the simultaneous effects of miR-16 on multiple myofibroblast-dependent signaling pathways, and in turn adjusting of the whole pathway–pathway network through “crosstalk63”.
HSC-to-myofibroblast transdifferentiation takes place upon the cooperation of multiple signaling pathways. Signaling pathways of TGF-β, Phosphatidylinositol-3-kinase/Akt, mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK), etc. contribute to the imbalance of ECM synthesis/degradation2,55,64–66. In addition, signaling pathways of Cell cycle, TGF-β, MAPK/ERK, etc. enable the over-proliferation of myofibroblasts2,58,61,67. Abnormal apoptosis is attributed to signaling pathways of Wnt, p53, and Apoptosis, etc.2,13,68,69 The chemotactic and cell–ECM interaction properties correlated with signaling pathways of Toll-like receptor and Focal adhesion2,60,70. This redundant and interactive network of signaling pathways makes it difficult to reverse the phenotype of myofibroblasts by traditional one-signaling pathway interference. miR-16 restoration, however, abolished the fibrosis-inducing characteristics, and reactivated the adipogenic property by signaling pathways of TGF-β and Wnt, respectively, in an integrated way. Hence, a dramatic transition from myofibroblasts to HSC-like phenotype was finally obtained, resulting in the substantial resolution of liver fibrosis.
On the other side, abrogation of miR-16 expression in HSCs by means of gene knockout or knockdown could be an alternative for revealing its pathophysiological effects. But primary HSCs experience spontaneous transdifferentiation (activation) toward myofibroblasts, with initial and permanent activation in succession, after their separation from the sinusoidal niche of liver. A progressive descending of miR-16 level occurs during the HSC-to-myofibroblast transdifferentiation13,71,72. Moreover, HSCs within this period demonstrate injury susceptibility to genetic engineering and lipofectamine toxicity73,74. Given the limitations that prevent HSCs from miR-16 knockout or knockdown, miR-16 inhibitor was employed in the present experiments so as to mimic, to some extent, the miRNA abrogation. In contrast to the regaining of HSC-like phenotype by miR-16 restoration, treatment of miR-16 inhibitor retained the myofibroblast phenotype with reactivation of miR-16’s targets and related signaling pathways.
In conclusion, miR-16 functions by synergistically targeting a set of signaling pathways essential for myofibroblasts, such as Wnt and TGF-β, thereby globally altering the transcriptome, reversing fibrosis-related phenotypes, and inducing the resolution of liver fibrosis. Therefore, miR-16 is involved in cell fate determination in myofibroblast and may be a novel therapeutic target for hepatic fibrosis.
Supplementary information
Acknowledgements
This work is supported by National Key R&D Program of China (2017YFC0908903) and National Natural Science Foundation of China (81270492, 81470859, and 81570544). We thank Chen Dai of Frice Bioinformatics Company, Li Qi of Genminix Information Company for technical assistance, and Laura E. Nagy of Case Western Reserve University for manuscript drafting.
Conflict of interest
The authors declare that they have no conflict of interest.
Footnotes
Edited by G. Blandino
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
These authors contributed equally: Qin Pan, Can-Jie Guo, Qing-Yang Xu
Supplementary information
Supplementary Information accompanies this paper at (10.1038/s41419-020-02832-z).
References
- 1.Trautwein C, Friedman SL, Schuppan D, Pinzani M. Hepatic fibrosis: concept to treatment. J. Hepatol. 2015;62:S15–S24. doi: 10.1016/j.jhep.2015.02.039. [DOI] [PubMed] [Google Scholar]
- 2.Tsuchida T, Friedman SL. Mechanisms of hepatic stellate cell activation. Nat. Rev. Gastroenterol. Hepatol. 2017;14:397–411. doi: 10.1038/nrgastro.2017.38. [DOI] [PubMed] [Google Scholar]
- 3.Tang Y. Curcumin targets multiple pathways to halt hepatic stellate cell activation: updated mechanisms in vitro and in vivo. Digest. Dis. Sci. 2015;60:1554–1564. doi: 10.1007/s10620-014-3487-6. [DOI] [PubMed] [Google Scholar]
- 4.Djuranovic S, Nahvi A, Green R. miRNA-mediated gene silencing by translational repression followed by mRNA deadenylation and decay. Science. 2012;336:237–240. doi: 10.1126/science.1215691. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Choi, Y. J. et al. Deficiency of microRNA miR-34a expands cell fate potential in pluripotent stem cells. Science355, 10.1126/science.aag1927 (2017). [DOI] [PMC free article] [PubMed]
- 6.Cordes KR, et al. MiR-145 and miR-143 regulate smooth muscle cell fate and plasticity. Nature. 2009;460:705–710. doi: 10.1038/nature08195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Boettger T, et al. Acquisition of the contractile phenotype by murine arterial smooth muscle cells depends on the Mir143/145 gene cluster. J. Clin. Investig. 2009;119:2634–2647. doi: 10.1172/JCI38864. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Inose H, et al. A microRNA regulatory mechanism of osteoblast differentiation. Proc. Natl Acad. Sci. USA. 2009;106:20794–20799. doi: 10.1073/pnas.0909311106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Le MTN, et al. MicroRNA-125b promotes neuronal differentiation in human cells by repressing multiple targets. Mol. Cell. Biol. 2009;29:5290–5305. doi: 10.1128/MCB.01694-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Calin GA, et al. A MicroRNA signature associated with prognosis and progression in chronic lymphocytic leukemia. N. Engl. J. Med. 2005;353:1793–1801. doi: 10.1056/NEJMoa050995. [DOI] [PubMed] [Google Scholar]
- 11.Cimmino A, et al. miR-15 and miR-16 induce apoptosis by targeting BCL2. Proc. Natl Acad. Sci. USA. 2005;102:13944–13949. doi: 10.1073/pnas.0506654102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Fong MY, et al. Breast-cancer-secreted miR-122 reprograms glucose metabolism in premetastatic niche to promote metastasis. Nat. Cell Biol. 2015;17:183–194. doi: 10.1038/ncb3094. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Guo CJ, Pan Q, Li DG, Sun H, Liu BW. miR-15b and miR-16 are implicated in activation of the rat hepatic stellate cell: An essential role for apoptosis. J. Hepatol. 2009;50:766–778. doi: 10.1016/j.jhep.2008.11.025. [DOI] [PubMed] [Google Scholar]
- 14.Taulli R, et al. The muscle-specific microRNA miR-206 blocks human rhabdomyosarcoma growth in xenotransplanted mice by promoting myogenic differentiation. J. Clin. Investig. 2009;119:2366–2378. doi: 10.1172/JCI38075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Bouhallier F, et al. Role of miR-34c microRNA in the late steps of spermatogenesis. RNA. 2010;16:720–731. doi: 10.1261/rna.1963810. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Lim LP, et al. Microarray analysis shows that some microRNAs downregulate large numbers of-target mRNAs. Nature. 2005;433:769–773. doi: 10.1038/nature03315. [DOI] [PubMed] [Google Scholar]
- 17.Rogler CE, et al. MicroRNA-23b cluster microRNAs regulate transforming growth factor-beta/bone morphogenetic protein signaling and liver stem cell differentiation by targeting Smads. Hepatology. 2009;50:575–584. doi: 10.1002/hep.22982. [DOI] [PubMed] [Google Scholar]
- 18.Jin, H. Y. et al. Differential Sensitivity of Target Genes to Translational Repression by miR-17~92. PLoS Genet.13, 10.1371/journal.pgen.1006623 (2017). [DOI] [PMC free article] [PubMed]
- 19.Vigorito E, et al. microRNA-155 regulates the generation of immunoglobulin class-switched plasma cells. Immunity. 2007;27:847–859. doi: 10.1016/j.immuni.2007.10.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Seok H, Ham J, Jang ES, Chi SW. MicroRNA target recognition: Insights from transcriptome-wide non-canonical interactions. Mol. Cells. 2016;39:375–381. doi: 10.14348/molcells.2016.0013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Hanlon, K., Rudin, C. E. & Harries, L. W. Investigating the targets of MIR-15a and MIR-16-1 in patients with chronic lymphocytic leukemia (CLL). PLoS ONE4, 10.1371/journal.pone.0007169 (2009). [DOI] [PMC free article] [PubMed]
- 22.Chen Z, et al. MicroRNA-181b suppresses TAG via target IRS2 and regulating multiple genes in the Hippo pathway. Exp. Cell Res. 2016;348:66–74. doi: 10.1016/j.yexcr.2016.09.004. [DOI] [PubMed] [Google Scholar]
- 23.Surdziel E, et al. Enforced expression of miR-125b affects myelopoiesis by targeting multiple signaling pathways. Blood. 2011;117:4338–4348. doi: 10.1182/blood-2010-06-289058. [DOI] [PubMed] [Google Scholar]
- 24.Fukaya T, Tomari Y. MicroRNAs mediate gene silencing via multiple different pathways in Drosophila. Mol. Cell. 2012;48:825–836. doi: 10.1016/j.molcel.2012.09.024. [DOI] [PubMed] [Google Scholar]
- 25.Popov Y, et al. Halofuginone induces matrix metalloproteinases in rat hepatic stellate cells via activation of p38 and NFκB. J. Biol. Chem. 2006;281:15090–15098. doi: 10.1074/jbc.M600030200. [DOI] [PubMed] [Google Scholar]
- 26.Montosi G, Garuti C, Iannone A, Pietrangelo A. Spatial and temporal dynamics of hepatic stellate cell activation during oxidant-stress-induced fibrogenesis. Am. J. Pathol. 1998;152:1319–1326. [PMC free article] [PubMed] [Google Scholar]
- 27.Dennis G, Jr, et al. DAVID: database for annotation, visualization, and integrated discovery. Genome Biol. 2003;4:P3. [PubMed] [Google Scholar]
- 28.Hu B, et al. CCAAT/Enhancer-binding protein β isoforms and the regulation of α-smooth muscle actin gene expression by IL-1β. J. Immunol. 2004;173:4661–4668. doi: 10.4049/jimmunol.173.7.4661. [DOI] [PubMed] [Google Scholar]
- 29.Li G, et al. Inhibition of connective tissue growth factor by siRNA prevents liver fibrosis in rats. J. Gene Med. 2006;8:889–900. doi: 10.1002/jgm.894. [DOI] [PubMed] [Google Scholar]
- 30.Jia X, et al. MiR-16 regulates mouse peritoneal macrophage polarization and affects T-cell activation. J. Cell Mol. Med. 2016;20:1898–1907. doi: 10.1111/jcmm.12882. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Zurawek D, et al. Time-dependent miR-16 serum fluctuations together with reciprocal changes in the expression level of miR-16 in mesocortical circuit contribute to stress resilient phenotype in chronic mild stress - An animal model of depression. Eur. Neuropsychopharmacol. 2016;26:23–36. doi: 10.1016/j.euroneuro.2015.11.013. [DOI] [PubMed] [Google Scholar]
- 32.She H, Xiong S, Hazra S, Tsukamoto H. Adipogenic transcriptional regulation of hepatic stellate cells. J. Biol. Chem. 2005;280:4959–4967. doi: 10.1074/jbc.M410078200. [DOI] [PubMed] [Google Scholar]
- 33.Panebianco C, Oben JA, Vinciguerra M, Pazienza V. Senescence in hepatic stellate cells as a mechanism of liver fibrosis reversal: a putative synergy between retinoic acid and PPAR-gamma signalings. Clin. Exp. Med. 2017;17:269–280. doi: 10.1007/s10238-016-0438-x. [DOI] [PubMed] [Google Scholar]
- 34.Sharvit, E., Abramovitch, S., Reif, S. & Bruck, R. Amplified Inhibition of Stellate Cell Activation Pathways by PPAR-γ, RAR and RXR Agonists. PLoS ONE8, 10.1371/journal.pone.0076541 (2013). [DOI] [PMC free article] [PubMed]
- 35.Li M, et al. SIRT1 antagonizes liver fibrosis by blocking hepatic stellate cell activation in mice. FASEB J. 2018;32:500–511. doi: 10.1096/fj.201700612R. [DOI] [PubMed] [Google Scholar]
- 36.Ding D, et al. Trichostatin A inhibits the activation of hepatic stellate cells by increasing C/EBP-α acetylation in vivo and in vitro. Sci. Rep. 2018;8:4395. doi: 10.1038/s41598-018-22662-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Tao LL, et al. TSA increases C/EBP-α expression by increasing its lysine acetylation in hepatic stellate cells. Mol. Med. Rep. 2017;16:6088–6093. doi: 10.3892/mmr.2017.7358. [DOI] [PubMed] [Google Scholar]
- 38.Yi SH, Zhang Y, Tang D, Zhu L. Mechanical force and tensile strain activated hepatic stellate cells and inhibited retinol metabolism. Biotechnol. Lett. 2015;37:1141–1152. doi: 10.1007/s10529-015-1785-5. [DOI] [PubMed] [Google Scholar]
- 39.Choi S, et al. Recombinant fusion protein of albumin-retinol binding protein inactivates stellate cells. Biochem. Biophys. Res. Commun. 2012;418:191–197. doi: 10.1016/j.bbrc.2012.01.012. [DOI] [PubMed] [Google Scholar]
- 40.Raghavan CT, et al. AGEs in human lens capsule promote the TGFβ2-mediated EMT of lens epithelial cells: Implications for age-associated fibrosis. Aging Cell. 2016;15:465–476. doi: 10.1111/acel.12450. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Bultmann-Mellin I, et al. Role of LTBP4 in alveolarization, angiogenesis, and fibrosis in lungs. Am. J. Physiol. Lung Cell. Mol. Physiol. 2017;313:L687–L698. doi: 10.1152/ajplung.00031.2017. [DOI] [PubMed] [Google Scholar]
- 42.Tao R, et al. MicroRNA-29b-3p prevents Schistosoma japonicum-induced liver fibrosis by targeting COL1A1 and COL3A1. J. Cell. Biochem. 2018;119:3199–3209. doi: 10.1002/jcb.26475. [DOI] [PubMed] [Google Scholar]
- 43.Frese, J. et al. Kidney injury by variants in the COL4A5 gene aggravated by polymorphisms in slit diaphragm genes causes focal segmental glomerulosclerosis. Int. J. Mol. Sci.20, 10.3390/ijms20030519 (2019). [DOI] [PMC free article] [PubMed]
- 44.Piccoli MT, et al. Inhibition of the cardiac fibroblast-enriched lncRNA Meg3 prevents cardiac fibrosis and diastolic dysfunction. Circ. Res. 2017;121:575–583. doi: 10.1161/CIRCRESAHA.117.310624. [DOI] [PubMed] [Google Scholar]
- 45.Li N, et al. p27 inhibits CDK6/CCND1 complex formation resulting in cell cycle arrest and inhibition of cell proliferation. Cell Cycle. 2018;17:2335–2348. doi: 10.1080/15384101.2018.1526598. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Si Y, et al. Fisetin decreases TET1 activity and CCNY/CDK16 promoter 5hmC levels to inhibit the proliferation and invasion of renal cancer stem cell. J. Cell. Mol. Med. 2019;23:1095–1105. doi: 10.1111/jcmm.14010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Shang G, Ma X, Lv G. Cell division cycle 20 promotes cell proliferation and invasion and inhibits apoptosis in osteosarcoma cells. Cell Cycle. 2018;17:43–52. doi: 10.1080/15384101.2017.1387700. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Choudhari R, et al. Redundant and nonredundant roles for Cdc42 and Rac1 in lymphomas developed in NPM-ALK transgenic mice. Blood. 2016;127:1297–1306. doi: 10.1182/blood-2015-11-683052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Farrugia AJ, Calvo F. The Borg family of Cdc42 effector proteins Cdc42EP1-5. Biochem. Soc. Trans. 2016;44:1709–1716. doi: 10.1042/BST20160219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Cohen S, et al. Cdc42 regulates the cellular localization of Cdc42ep1 in controlling neural crest cell migration. J. Mol. Cell Biol. 2018;10:376–387. doi: 10.1093/jmcb/mjx044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Nayak, G. et al. Developmental vascular regression is regulated by a wnt/β-catenin, myc and cdkn1a pathway that controls cell proliferation and cell death. Development (Cambridge)145, 10.1242/dev.154898 (2018). [DOI] [PMC free article] [PubMed]
- 52.Jensen SA, et al. Bcl2L13 is a ceramide synthase inhibitor in glioblastoma. Proc. Natl Acad. Sci. USA. 2014;111:5682–5687. doi: 10.1073/pnas.1316700111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Camargo A, et al. Dietary fat may modulate adipose tissue homeostasis through the processes of autophagy and apoptosis. Eur. J. Nutr. 2017;56:1621–1628. doi: 10.1007/s00394-016-1208-y. [DOI] [PubMed] [Google Scholar]
- 54.Sharifi M, Moridnia A. Apoptosis-inducing and antiproliferative effect by inhibition of miR-182-5p through the regulation of CASP9 expression in human breast cancer. Cancer Gene Ther. 2017;24:75–82. doi: 10.1038/cgt.2016.79. [DOI] [PubMed] [Google Scholar]
- 55.Bonci D, et al. The miR-15a-miR-16-1 cluster controls prostate cancer by targeting multiple oncogenic activities. Nat. Med. 2008;14:1271–1277. doi: 10.1038/nm.1880. [DOI] [PubMed] [Google Scholar]
- 56.Perumal N, Perumal M, Halagowder D, Sivasithamparam N. Morin attenuates diethylnitrosamine-induced rat liver fibrosis and hepatic stellate cell activation by co-ordinated regulation of Hippo/Yap and TGF-β1/Smad signaling. Biochimie. 2017;140:10–19. doi: 10.1016/j.biochi.2017.05.017. [DOI] [PubMed] [Google Scholar]
- 57.Sun H, et al. Oligo-peptide I-C-F-6 inhibits hepatic stellate cell activation and ameliorates CCl4-induced liver fibrosis by suppressing NF-κB signaling and Wnt/β-catenin signaling. J. Pharmacol. Sci. 2018;136:133–141. doi: 10.1016/j.jphs.2018.01.003. [DOI] [PubMed] [Google Scholar]
- 58.Ezhilarasan D, et al. Silibinin induces hepatic stellate cell cycle arrest via enhancing p53/p27 and inhibiting Akt downstream signaling protein expression. Hepatobiliary Pancreat. Dis. Int. 2017;16:80–87. doi: 10.1016/s1499-3872(16)60166-2. [DOI] [PubMed] [Google Scholar]
- 59.Nishizawa H, et al. IGF-I induces senescence of hepatic stellate cells and limits fibrosis in a p53-dependent manner. Sci. Rep. 2016;6:34605. doi: 10.1038/srep34605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Zhao XK, et al. Focal adhesion kinase regulates hepatic stellate cell activation and liver fibrosis. Sci. Rep. 2017;7:4032. doi: 10.1038/s41598-017-04317-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Shah R, et al. TGF-β1 up-regulates the expression of PDGF-β receptor mRNA and induces a delayed PI3K-, AKT-, and p70S6K-dependent proliferative response in activated hepatic stellate cells. Alcohol. Clin. Exp. Res. 2013;37:1838–1848. doi: 10.1111/acer.12167. [DOI] [PubMed] [Google Scholar]
- 62.Ross SE, et al. Inhibition of adipogenesis by Wnt signaling. Science. 2000;289:950–953. doi: 10.1126/science.289.5481.950. [DOI] [PubMed] [Google Scholar]
- 63.Carthy, J. M., Garmaroudi, F. S., Luo, Z. & McManus, B. M. Wnt3a induces myofibroblast differentiation by upregulating TGF-β signaling through SMAD2 in a β-catenin-dependent manner. PLoS ONE6, 10.1371/journal.pone.0019809 (2011). [DOI] [PMC free article] [PubMed]
- 64.Iwasaki A, et al. Molecular mechanism responsible for fibronectin-controlled alterations in matrix stiffness in advanced chronic liver fibrogenesis. J. Biol. Chem. 2016;291:72–88. doi: 10.1074/jbc.M115.691519. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Wei L, et al. Asiatic acid attenuates CCl4-induced liver fibrosis in rats by regulating the PI3K/AKT/mTOR and Bcl-2/Bax signaling pathways. Int. Immunopharmacol. 2018;60:1–8. doi: 10.1016/j.intimp.2018.04.016. [DOI] [PubMed] [Google Scholar]
- 66.Guo Y, et al. Insulin-like growth factor binding protein-related protein 1 (IGFBPrP1) contributes to liver inflammation and fibrosis via activation of the ERK1/2 pathway. Hepatol. Int. 2015;9:130–141. doi: 10.1007/s12072-014-9578-9. [DOI] [PubMed] [Google Scholar]
- 67.Sun WY, et al. Depletion of β-arrestin2 in hepatic stellate cells reduces cell proliferation via ERK pathway. J. Cell. Biochem. 2013;114:1153–1162. doi: 10.1002/jcb.24458. [DOI] [PubMed] [Google Scholar]
- 68.Cai SP, et al. Transmembrane protein 88 attenuates liver fibrosis by promoting apoptosis and reversion of activated hepatic stellate cells. Mol. Immunol. 2016;80:58–67. doi: 10.1016/j.molimm.2016.11.002. [DOI] [PubMed] [Google Scholar]
- 69.Huang YH, et al. Interleukin-10 promotes primary rat hepatic stellate cell senescence by upregulating the expression levels of p53 and p21. Mol. Med. Rep. 2018;17:5700–5707. doi: 10.3892/mmr.2018.8592. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Kumar S, Wang J, Shanmukhappa SK, Gandhi CR. Toll-like receptor 4–independent carbon tetrachloride–induced fibrosis and lipopolysaccharide-induced acute liver injury in mice: role of hepatic stellate cells. Am. J. Pathol. 2017;187:1356–1367. doi: 10.1016/j.ajpath.2017.01.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Kim KM, et al. Gα12 overexpression induced by miR16 dysregulation contributes to liver fibrosis by promoting autophagy in hepatic stellate cells. J. Hepatol. 2018;68:493–504. doi: 10.1016/j.jhep.2017.10.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Guo CJ, et al. Changes in microRNAs associated with hepatic stellate cell activation status identify signaling pathways. FEBS J. 2009;276:5163–5176. doi: 10.1111/j.1742-4658.2009.07213.x. [DOI] [PubMed] [Google Scholar]
- 73.Friedman SL. Stellate cells: a moving target in hepatic fibrogenesis. Hepatology. 2004;40:1041–1043. doi: 10.1002/hep.20476. [DOI] [PubMed] [Google Scholar]
- 74.Friedman SL, Bansal MB. Reversal of hepatic fibrosis - fact or fantasy? Hepatology. 2006;43:S82–S88. doi: 10.1002/hep.20974. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.