Clin Mol Hepatol > Volume 31(3); 2025 > Article
Kim, Park, Park, Kim, Lim, Lee, Bae, and Kim: Modulation of phosphatase of regenerating liver-1 within placental mesenchymal stem cells instigates the transition between epithelial-to-mesenchymal transition and mesenchymal-to-epithelial transition subsequent to hepatic fibrosis

ABSTRACT

Background/Aims

Epithelial-to-mesenchymal transition (EMT) plays a crucial role in hepatic fibrogenesis and liver repair in chronic liver disease. Our research highlights the antifibrotic potential of placenta-derived mesenchymal stem cells (PD-MSCs) and the role of phosphatase of regenerating liver-1 (PRL-1) in promoting liver regeneration.

Methods

We evaluated the efficacy of PD-MSCs overexpressing PRL-1 (PD-MSCsPRL-1) in a bile duct ligationinduced rat injury model, focusing on their ability to regulate EMT.

Results

PD-MSCsPRL-1 significantly reduced mesenchymal markers by downregulating TGFB1/SMAD2, outperforming naïve PD-MSCs. The transplantation of PD-MSCsPRL-1 enhanced BMP7/SMAD1/5 expression, promoting epithelial marker expression and stimulating BMP7 within hepatocytes, modulating downstream SMAD signaling. Importantly, further validation confirmed that PRL-1 directly interacts with BMP7 in hepatocytes.

Conclusions

PRL-1 expression in PD-MSCsPRL-1 restores TGFB1/BMP7 balance, promoting hepatic regeneration through mesenchymal-to-epithelial transition. These findings highlight the therapeutic potential of engineered MSCs for liver disease and suggest innovative strategies for future stem cell therapies.

Graphical Abstract

INTRODUCTION

Epithelial-to-mesenchymal transition (EMT) is essential in development, tissue repair, and cancer progression [1]. EMT enables epithelial cells to lose polarity and gain mesenchymal traits, enhancing migration, invasiveness, extracellular matrix (ECM) production, and apoptosis resistance [2]. In chronic liver diseases including cirrhosis, EMT sustains wound-healing responses, driving fibrotic pathways and regenerative nodule formation. Liver cirrhosis involves ECM deposition and hepatic stellate cell (HSC) activation, influenced by transforming growth factor beta 1 (TGFB1) and SMAD signaling [3]. These signals promote EMT in hepatic cells, marked by loss of epithelial markers and increased fibrotic markers such as α-smooth muscle actin and collagen, mediated through SMAD signaling pathways [4]. EMT has been identified as a key contributor to fibrogenesis, and reversing EMT could help restore liver function in chronic liver diseases [5].
Recently, mesenchymal stem cell (MSC) therapy has shown potential in liver fibrosis treatment due to their ability to promote tissue remodeling, secrete anti-inflammatory cytokines, and differentiate into hepatocyte-like cells [6]. In a prior study, we reported that MSCs derived from normal term placentas exhibited the capacity to enhance hepatic function via systemic pathways, these mechanisms encompassed anti-inflammatory, anti-fibrotic responses. However, naïve MSC therapy has exhibited short-term efficacy, underscoring its limited capacity for sustained amelioration of pathological conditions [7,8].
To address this limitation, we developed placenta-derived MSCs engineered to overexpress phosphatase of regener-ating liver-1 (PRL-1; PD-MSCsPRL-1) to enhance their intrinsic therapeutic potential, including homing to target tissues, migration, and survival [9]. PRL-1, a protein tyrosine phosphatase, is key in cellular proliferation and tissue regeneration. PRL-1 was first identified in regenerating rat liver and has been shown to maintain epithelial differentiation and promote cell cycle progression [10]. However, PRL-1 overexpression has also been linked to EMT and cancer progression, highlighting its dual roles in different cellular contexts [11]. Unlike liver cancer, where cells proliferate aberrantly to form tumors, chronic liver disease is characterized by a pronounced impairment of cellular proliferative capacity. Our previous research demonstrated that PD-MSCsPRL-1 significantly enhanced liver regeneration by metabolic alteration in a rat model of bile duct ligation (BDL) rather than that of naïve MSCs [12]. While this demonstrated the therapeutic potential of PD-MSCsPRL-1 in liver regeneration, their mechanical role in promoting mesenchymal-to-epithelial transition (MET)—the reverse process of EMT—within fibrotic liver conditions remains unexplored.
Given the pivotal role of MET in tissue repair and its potential to reverse fibrosis, this study investigates the effects of PD-MSCsPRL-1 on MET progression in a rat model of BDL-induced liver fibrosis. By examining how PD-MSCsPRL-1 modulate MET, we aim to demonstrate their ability to overcome the limitations of naïve MSCs by leveraging EMT-related mechanisms. Our findings could substantiate PD-MSCsPRL-1 as a promising approach for not only mitigating fibrosis but also promoting epithelial restoration, thereby facilitating normal liver function recovery in fibrotic conditions. This work may contribute to translational research on stem cell therapies for chronic liver diseases.

MATERIALS AND METHODS

Cell culture

The preparation of PD-MSCs was approved by the CHA Gangnam Medical Center Institutional Review Board (Seoul, Korea, No.: IRB 07-18) and isolated as previously described [13]. WB-F344 (a diploid epithelial cell line from normal adult rat liver with phenotypic properties of “oval” cells) and T-HSC/Cl-6 (rat HSCs) were cultured in α-MEM (HyClone, Logan, UT, USA) with 10% FBS (Gibco, Carlsbad, CA, USA) and 1% penicillin/streptomycin (P/S, Gibco). Primary hepatocytes from 7-week-old male Sprague–Dawley rats (Orient Bio Inc., Seongnam, Korea) were isolated via a two-step pronase/collagenase perfusion and cultured in William’s E medium (Sigma-Aldrich, St. Louis, MO, USA) with 10% FBS (Gibco), 1% P/S (Gibco), and 4 mM L-glutamine (Gibco). All cells were cultured at 37°C in 5% CO2. To analyze EMT induction and its reversal, each cell was treated with TGFB1 (2 ng/mL, PeproTech, Rocky Hill, NJ, USA) for 48 and 72 hours. Cells were then cocultured with PD-MSCs or PD-MSCsPRL-1 (5×10³ cells/cm²) in Transwell inserts (8-μm pore, Corning, NY, USA) using α-MEM (HyClone) with 10% FBS (Gibco) and 1% P/S (Gibco) for 24 hours.

Gene transfection and RNA interference

To induce overexpression in PD-MSCs, a human PRL-1 plasmid was purchased from Origene Inc. (Rockville, MD, USA) and transfected into naïve PD-MSCs using the 4D Nucleofector™ system (Lonza, Basel, Switzerland), as previously described [14]. For gene knockdown, small interfering (si) RNA targeting PRL-1 and BMP7, along with scrambled RNA, were obtained from Integrated DNA Technologies (San Jose, CA, USA). Cells at 80% confluence were transfected with 50 nM siRNA using Lipofectamine RNAiMAX following the manufacturer’s instructions (Invitrogen, Carlsbad, CA, USA).

Proteome profiler human cytokine array

A cytokine antibody array analysis was performed using the Proteome Profiler Human XL Cytokine Array Kit (R&D Systems, Minneapolis, MN, USA). In brief, the supernatants from cultured PD-MSCs and PD-MSCsPRL-1 were collected. The array procedure strictly adhered to the manufacturer’s instructions. Each membrane was equipped with 105 cytokine antibodies, each of which was spotted in duplicate.

Animal experiments

Animal protocols were approved by the CHA University Animal Care Committee (IACUC-190048). Male Sprague–Dawley rats (7 week-old, 200 g; Orient Bio Inc.) were maintained under specific pathogen-free conditions with a 12-hour light/dark cycle at 22–25°C. Af ter 1 week of acclimatization, liver cirrhosis was induced by BDL for 10 days [15]. Rats were randomly divided into groups: BDL-induced non-transplanted (BDL NTx; n=20) and sham controls (Con; n=6). PKH67 (Sigma-Aldrich)-labeled PD-MSCs (n=20) and PD-MSCsPRL-1 (n=20) were transplanted intravenously (2×106 cells/animal). Rats were sacrificed at 1, 2, 3, and 5 weeks for analysis.

Biochemical analysis

The levels of aspartate aminotransferase (AST), alanine aminotransferase (ALT), total bilirubin, and albumin were assessed in individual serum samples from each group (n=5). Each serum sample was subjected to analysis upon test request (Southeast Medi-Chem Institute, Busan, Korea).

Liver histology and immunostainings

For histopathological analysis, liver specimens (n=5/group) were fixed in 10% neutral buffered formalin, embedded in paraffin, and stained with hematoxylin & eosin (H&E), Masson’s trichrome, and Sirius red. For immunohistochemistry, liver sections were deparaffinized, hydrated, and treated with 3% hydrogen peroxide in methanol. After antigen retrieval, specimens were incubated in Protein Block solution (DAKO, Santa Clara, CA, USA), followed by antibodies against PCNA (1:50, Santa Cruz Biotechnology Inc., Dallas, Texas, USA), PRL-1 (1:200, Abcam, Cambridge, UK), and BMP7 (1:100, Abcam) at 4°C overnight. 3,3-Diaminobenzidine (EnVision Systems, Santa Clara, CA, USA) was used to generate chromatic signals, and the samples were counterstained with Mayer’s hematoxylin (DAKO). Whole liver tissue images were analyzed with a digital slide scanner (3DHISTECH Ltd., Budapest, Hungary). For immunofluorescence (IF), primary antibodies against E-cadherin (1:200, Cell Signaling Technology), BMP7 (1:200, Abcam), p-SMAD2 (1:200, Cell Signaling Technology), Ki -67 (DAKO), and HNF4A (Novus Biologicals, Littleton, CO, USA) were used in diluent solution (DAKO). 4’,6-Diamidino-2-phenylindole (DAPI) (Invitrogen) was used as a counterstain and slides were visualized by confocal microscopy (LSM 700). Images were analyzed with ZEN blue software (Zeiss).

Co-immunoprecipitation and immunoblotting

Liver tissues and cells were lysed with Pierce immunoprecipitation (IP) buffer (Thermo Fisher, Waltham, MA, USA) for Co-IP and RIPA buffer (Sigma-Aldrich) for immunoblotting containing protease and phosphatase inhibitors. For Co-IP, lysates were incubated with 2 μg of anti-PRL-1 (Abcam) overnight with gentle rotation. Protein G agarose (Thermo Fisher) was added to the antibody-labeled lysates, followed by the mixture bieng incubated for 2 hours at 4°C with gentle rotation. The bound proteins were eluted and immunoblotting was performed to detect BMP7 using anti-BMP7 (Abcam). For immunoblotting, total protein or nuclear-cytoplasmic fractioned (Thermo Fisher) samples (40 μg) from tissues, cells, nuclei, and cytoplasm were separated by SDS-PAGE. Primary antibodies against ALB, BCL2, COL1A1 (Novus Biologicals), ACTA2 (Sigma-Aldrich), BAX, VIM, SNAI1, p-SMAD1/5, GP130 (Santa Cruz Biotechnology), PARP, total/p-SMAD2, TJP, CDH1, total/p-STAT3, interleukin (IL)-6, CDK4, CCND1, CASPASE-3 (Cell Signaling Technology, Danvers, MA, USA), PRL-1, BMP7, and total SMAD1/5 (Abcam) were used. GAPDH (AbFrontier, Seoul, Korea) and LMNB1 (Cell Signaling Technology) were loading controls for cytoplasmic and nuclear proteins. HRP-conjugated secondary antibodies were applied for 1 hour incubation and chemiluminescence detection.

Quantitative real-time polymerase chain reaction

Total RNA was extracted from liver tissues from each group or cells with TRIzol LS reagent (Invitrogen), and Quantitative real-time polymerase chain reaction was performed using SYBR Green PCR Master Mix (Applied Biosystems, Waltham, MA, USA) on a CFX Connect™ Real-Time System (Bio-Rad, Hercules, CA, USA) as previously described [16]. The primers are listed in the Supplementary Table 1.

Enzyme-linked immunosorbent assay

Exogenous levels were measured using Enzyme-linked immunosorbent assay (ELISA) kits using rat IL-6 and rat IL-10 (R&D Systems), rat TGFB1, rat/human BMP7 (Abcam) in rat serum, and human PRL-1 (MyBioSource, Inc., San Diego, CA, USA) in cell culture supernatant, following the manufacturer’s instructions. The absorbance was measured using an Epoch microplate reader (BioTek, Winooski, VT, USA) at 450 nm. The experiments were performed in triplicate.

Wound healing assay

WB-F344 were cultured in 6-well plates at a density of 2×105 cells/well. The following day, a sterile 1,000 μl tip was used to create a straight-line scratch across the monolayer and subsequently they were treated with TGFB1 (PeproTech) for 48 hours, followed by treatment with PRL-1 (Novus Biologicals) and Pentamidine (Sigma-Aldrich), which is a PRL-1 inhibitor, respectively. The extent of wound closure was quantified by measuring the gap width at identical locations using ImageJ software (NIH).

Statistical analysis

All data were analyzed using GraphPad Prism version 9.0 (GraphPad Software, San Diego, CA USA), and represented as means±standard deviations. Statistical analysis was performed using two-tailed unpaired Student’s t-test and one-way ANOVA, and the difference was considered significant when the P-value was less than 0.05. All data are representative of at least three independent experiments.

RESULTS

PD-MSCsPRL-1 mitigate hepatic fibrosis in a rat model subjected to bile duct ligation injury

In the BDL-induced cirrhosis model [15], rats were divided into four groups: BDL-induced nontransplantation (BDL NTx), naïve PD-MSCs, PD-MSCsPRL-1 transplantation, and Con (Fig. 1A). Prior studies underscored the superior antifibrotic efficacy of PD-MSCsPRL-1 transplantation, mediated by mitochondrial activation [12]. Histopathological evaluation revealed an elevated biliary ductal cell count in the BDL group, fostering tissue restitution while exacerbating fibrogenesis, inversely associated with survival rates (Fig. 1B). Collagen deposition was markedly augmented in the BDL NTx group compared to controls, whereas transplantation groups, particularly PD-MSCsPRL-1, demonstrated substantial fibrosis attenuation, corroborated by Sirius red and Masson’s trichrome staining (Fig. 1C). Fibrotic markers COL1A1 and ACTA2 were significantly elevated in the BDL group but reduced in both transplantation groups, with PD-MSCsPRL-1 showing greater reductions (Fig. 1D). To assess the recuperation of hepatic functions facilitated by transplantation, measurements of ALT, AST, and total bilirubin levels were conducted in the serum of rats. Liver function tests revealed that PD-MSCsPRL-1 significantly decreased ALT, AST, and total bilirubin levels compared to naïve PD-MSCs (Fig. 1E). Additionally, PD-MSCsPRL-1 increased IL-10 levels, indicating an enhanced anti-inflammatory response (Fig. 1F). These data demonstrate that PD-MSCsPRL-1 transplantation effectively mitigated hepatic fibrosis and enhanced hepatic function in a rat model subjected to BDL, as compared to PD-MSCs transplantation.

Transplantation of PD-MSCsPRL-1 modulates cellular survival and apoptosis in rats afflicted with BDL

We further analyzed secreted factors by cytokine array in the supernatant from PD-MSCsPRL-1 to demonstrate pathways associated with liver regeneration. Upregulated factors included FGF-2, GDF-15, HGF, and IL-6, while downregulated factors included DKK-1 and BDNF (Fig. 2A). To investigate hepatic regeneration, we focused on the IL-6/STAT3 pathway, known for its key role in liver regeneration [17]. IL-6 levels were significantly altered at 1 and 2 weeks post-PD-MSCsPRL-1 transplantation (Fig. 2B), with activation of IL-6, GP130, and p-STAT3 observed up to 3 weeks. A strong correlation (R=0.5852) was found between IL-6 and p-STAT3 levels (Fig. 2C). Serum and endogenous albumin levels were elevated in the PD-MSCsPRL-1 group compared to naïve. We further examined apoptosis and cell proliferation markers in liver tissues. BCL2 expression was reduced in the BDL-induced NTx group but elevated in both transplantation groups, with PD-MSCsPRL-1 showing higher levels at 3 weeks. Proapoptotic markers c-PARP and c-CASPASE-3 were reduced in both transplantation groups, with a greater reduction in the PD-MSCsPRL-1 group compared to PD-MSCs. Additionally, PD-MSCsPRL-1 significantly suppressed BAX expression, indicating a reduction in apoptosis (Fig. 2D). In our earlier findings, PD-MSCs transplantation in a rat model of CCl4-induced liver injury resulted in elevated BAX levels compared to the BDL-induced NTx group, indicating the initiation of an autophagic mechanism [7]. Cell cycle markers CDK4 and CCND1 were consistently elevated in the PD-MSCsPRL-1 group across all weeks (Fig. 2E), and PCNA levels, assessed by immunohistochemistry, were also increased (Fig. 2F). Moreover, Ki-67 and HNF4A co-staining suppor ted a hepatocyte proliferation in WB-F344 cells, followed by pentamidine treatment , a pharmacological inhibitor of PRL-1 (Supplementary Fig. 1). These results suggest that PD-MSCsPRL-1 transplantation enhances liver regeneration by inhibiting apoptosis and promoting hepatocyte proliferation in the BDL rat model.

PD-MSCsPRL-1 suppress mesenchymal phenotype by repressing SMAD2 signaling in fibrotic rat liver

Mesenchymal phenotypes, such as fibrosis or cirrhosis in liver tissues, often escalate under injurious conditions via EMT [18]. To determine whether MSC transplantation affects EMT progression by modulating TGFB1/SMAD2 signaling in the liver, we first measured TGFB1 levels in rat serum. The PD-MSCsPRL-1 group showed a two-fold reduction in TGFB1 levels compared to the PD-MSCs group, with a strong positive correlation (R=0.6430, Fig. 3A). Liver tissue, consisting of hepatocytes and cholangiocytes, showed mesenchymal phenotypes near the central vein in BDL-induced rats (Fig. 3B). After transplantation, there was a significant reduction in EMT markers, including Tgfb1, vimentin (Vim), and Snai1, compared to the NTx group, with the PD-MSCsPRL-1 group showing the most pronounced decrease (Fig. 3C). Protein expression of VIM and SNAI1, and especially phosphorylated SMAD2 (p-SMAD2) both total and nuclear, was also substantially reduced following PD-MSCsPRL-1 transplantation (Fig. 3D). While p-SMAD2 localized in the cytoplasm of hepatocytes in normal liver tissue, it translocated to the nucleus in the BDL NTx group which was reversed by PD-MSCs and PD-MSCsPRL-1 transplantation (Fig. 3E). These findings indicate that PD-MSCsPRL-1 transplantation effectively suppresses EMT progression by modulating the TGFB1/SMAD2 signaling pathway in BDL-injured rat livers (Fig. 3F).

PD-MSCsPRL-1 induce BMP7 expression through SMAD1/5 signaling

EMT induced by TGFB1 can inhibit BMP signaling, while BMP7 promotes MET [19]. To evaluate the impact of PD-MSCsPRL-1 on BMP7 levels in the BDL rat model, we measured serum BMP7 levels and its corresponding mRNA expression (Fig. 4A). A strong positive correlation (R=0.8121) was found between serum and mRNA Bmp7 levels. Additionally, phosphorylated SMAD1/5 (p-SMAD1/5), a downstream mediator of BMP7, was activated in both transplantation groups (Fig. 4B). Immunohistochemical analysis revealed that BMP7 and PRL-1 were predominantly localized in hepatocytes rather than cholangiocytes in rat liver tissues, with the PD-MSCsPRL-1 group showing higher expression (R=0.7261) (Fig. 4C). In addition, the mRNA expression of epithelial markers (e.g., desmoplakin; Dsp, cadherin 1; Cdh1, and tight junction protein; Tjp1) was dramatically increased by PD-MSCsPRL-1 transplantation compared to PD-MSCs transplantation (Fig. 4D) as well as protein levels (TJP and CDH1). IF confirmed elevated BMP7 and CDH1 levels in hepatocytes following PD-MSCsPRL-1 transplantation (Fig. 4E). These findings suggest that PD-MSCsPRL-1 activates BMP7 signaling in hepatocytes of cirrhotic livers, reducing EMT via the SMAD1/5 pathway (Fig. 4F).

Coculturing with PD-MSCsPRL-1 retains the epithelial phenotype of rat liver epithelial cells

To induce the progression of EMT in primary rat epithelial hepatocytes, a treatment with TGFB1 (2 ng/mL) for 48 hours was executed. To induce EMT in primary rat epithelial hepatocytes, TGFB1 (2 ng/mL) was applied for 48 hours. Subsequently, cells were cocultured with PD-MSCs or PD-MSCsPRL-1 for 24 hours. Prior to experimentation, WB-F344 cells were pretreated with TGFB1 for 48 or 72 hours, resulting in a mesenchymal phenotype, confirmed by reduced Cdh1 and increased Vim and Snai1 expression (Supplementary Figs. 2, 3). TGFB1 treatment increased mesenchymal markers, but coculturing with PD-MSCs or PD-MSCsPRL-1 significantly reduced TGFB1 expression (Fig. 5A). There was a strong negative correlation between TGFB1 and BMP7 expression (R=–0.9461). PRL-1 overexpression in PD-MSCs remarkably increased BMP7 and CDH1 levels, while PRL-1 knockdown led to the loss of the epithelial phenotype and decreased BMP7/SMAD1/5 signaling in primary hepatocytes (Fig. 5B). In the control group, both BMP7 and TGFB1 were secreted at approximately 500–700 pg/mL, but TGFB1-treated hepatocytes showed BMP7 loss and TGFB1 gain (Fig. 5C). Coculture with PD-MSCs or PD-MSCsPRL-1 decreased mesenchymal markers (TGFB1, SNAI1, COL1A1) and increased epithelial markers (BMP7, DSP, CDH1) at both mRNA and protein levels (Fig. 5B, 5D). PRL-1 knockdown in PD-MSCs led to worsened EMT, indicated by increased p-SMAD2 activity in hepatocytes (Fig. 5E). These findings suggest that PD-MSCsPRL-1 coculture suppresses the mesenchymal phenotype by downregulating p-SMAD2 and promotes the epithelial phenotype by upregulating BMP7/SMAD1/5 signaling (Fig. 5F).

PRL-1 modulates BMP7 expression

Given the strong positive correlation between PRL-1 and BMP7 in liver tissues, our analysis aimed to identify the upstream regulator between these two factors. Initially, we confirmed elevated expressions of both PRL-1 and BMP7 in human PD-MSCsPRL-1. Knockdown of PRL-1 in PD-MSCs resulted in diminished expression levels of both BMP7 and PRL-1, whereas silencing BMP7 had no discernible effect on PRL-1 expression, indicating the regulatory role of PRL-1 in modulating BMP7 levels. Recombinant PRL-1 treatment was administered to naïve PD-MSCs, resulting in increased PRL-1 and BMP7 mRNA and protein levels (Fig. 6A, Supplementary Fig. 4A4C). Further validation by costaining and Co-IP assay confirmed the interaction between PRL-1 and BMP7 (Fig. 6B), not SMAD1/5. We conducted BMP7 knockdown in primary hepatocytes and modulated p-SMAD1/5 and p-SMAD2 expression (Fig. 6C). To substantiate the role of PRL-1, we observed that treatment with recombinant PRL-1 markedly suppressed cell migration in WB-F344, whereas the application of pentamidine exhibited an opposing effect by a wound healing assay (Fig. 6D). Importantly, PRL-1 treatment alone upregulated p-SMAD1/5 and downregulated p-SMAD2. Conversely, pentamidine increased p-SMAD2 expression (Fig. 6E). These findings suggest that PRL-1 directly regulates BMP7, and that BMP7, under the influence of PRL-1, modulates p-SMAD1/5 and p-SMAD2 activity in rat liver epithelial cells (Fig. 6F).

DISCUSSION

MSC therapy is emerging as a promising treatment for chronic liver diseases, offering benefits such as reducing inflammation, promoting tissue repair, and regenerating damaged liver tissue [20]. Current stem cell therapies, such as autologous MSC transplantation (Cellgram-LC by Pharmicell Co., Ltd.), have demonstrated efficacy in improving liver function and reducing fibrosis in clinical trials for alcoholic liver cirrhosis (ClinicalTrials.gov/NCT05093881) and allogenic UC-MSC therapy (Cl i n icalTr ials.gov/NCT01220492). These studies provide encouraging evidence of the safety and therapeutic potential of stem cell-based interventions over extended periods. Despite these advancements in MSC therapy, effective liver repair requires replacing dead hepatocytes and epithelial cells with healthy ones, a fundamental challenge that remains unresolved in chronic liver disease. In chronic liver disease, especially cirrhosis, parenchymal cells cannot regenerate to replace damaged cells due to persistent inflammation, fibrotic tissue deposition, and disrupted cellular signaling pathways [21]. One of the key mechanisms underlying this failure of regeneration is type 2 EMT, driven by the TGFB/SMAD signaling pathway, which plays a pivotal role in liver fibrosis and disease progression [22]. A prior study reported that exosomes from UC-MSCs effectively suppressed TGFB1 and N-cadherin (CDH2), markers of the mesenchymal phenotype, thus inhibiting EMT progression and protecting hepatocytes. While previous work showed that UCMSC-derived exosomes suppress EMT, the exact mechanism remains unclear [23].
Our study demonstrate that PD-MSCsPRL-1 transplantation was achieved through the reduction of mesenchymal markers via TGFB1/SMAD2 downregulation, while BMP7, a natural antagonist of TGFB1, activated SMAD1/5 signaling in conjunction with PD-MSCsPRL-1. Consistent with our findings, previous research suggests that recombinant BMP7 inhibits mesenchymal cell proliferation while promoting hepatocyte growth in CCl4-induced liver fibrosis models [24], reducing collagen deposition and TIMP2 expression, and restoring liver function [25]. The interaction between the TGFB1 and BMP pathways shows an inverse relationship in hepatic fibrosis development [26]. TGFB1 suppresses BMP2 expression in HSCs, while BMP2 counteracts TGFB1’s effects by reducing its ligand and receptor expression. This leads to a decrease in TGFB1-dependent p-SMAD3 signaling and EMT markers like ACTA2 and FN1 [27]. In animal models, recombinant human BMP7 significantly reduced collagen deposition and decreased COL1A1 expression in HSCs, protecting against p-SMAD2/3 activation [28]. BMP7, along with p-SMAD1/5/8, antagonizes TGFB1 signaling by competing with p-SMAD2/3 for DNA binding or co-factor recruitment [29]. This TGFB1-BMP interplay also impacts liver fibrogenesis and related conditions, such as metabolic dysfunction-associated fatty liver disease.
PRL-1, localized in hepatocytes, correlated positively with BMP7 after partial hepatectomy [30]. Importantly, we confirmed interaction between PRL-1 and BMP7 in hepatocytes. Recombinant PRL-1 reduced p-SMAD2 in TGFB1-induced hepatocytes, while PRL-1 inhibition decreased p-SMAD1/5 activity. Furthermore, we are actively exploring whether recombinant PRL-1 injection alone could offer significant efficacy in treating hepatic fibrosis. Human PRL-1 exhibits complete amino acid sequence identity with rat PRL-1 [31]. We are considering the necessity of preliminary findings on acute toxicity (half-life), repeated-dose toxicity, and carcinogenic and reproductive toxicity studies, in accordance with the nonclinical testing guidelines of the Ministry of Food and Drug Safety. We are also keen to investigate whether PRL-1 recombinant protein exhibits a homing capability analogous to that observed with MSC administration.
Our findings reveal that PD-MSCsPRL-1 transplantation ameliorates hepatic fibrosis and enhances liver functionality compared to PD-MSCs, offering promising potential for regenerative medicine and immunomodulatory therapies in future medical applications. Enhanced immunosuppressive and anti-inflammatory properties of PD-MSCsPRL-1 compared to unmodified MSCs suggest substantial therapeutic benefits for managing chronic inflammation and immunemediated damage, such as cirrhosis and other inflammatory pathologies. This advancement provides a novel approach to suppress deleterious immune responses while fostering tissue repair, addressing a critical need in immune-compromised patients. However, translating these preclinical outcomes into human applications entails significant hurdles, including the need to meticulously evaluate the long-term safety profile of PRL-1 overexpression due to potential oncogenicity and unforeseen cellular proliferation. Ensuring consistent therapeutic effects across diverse patient populations necessitates rigorous trials to confirm the reproducibility and stability of PD-MSCsPRL-1’s effects. Additionally, scalable production protocols under Good Manufacturing Practices (GMP) conditions are essential to maintain cellular integrity and phenotypic fidelity during in vitro expansion. The interaction of PD-MSCsPRL-1 with existing immunosuppressive regimens warrants further investigation, as adjunctive therapies may be required if their immunomodulatory properties prove insufficient. Collectively, while PD-MSCsPRL-1 represents an innovative therapeutic avenue, its clinical adaptation will require comprehensive validation and optimization through controlled clinical trials.
However, several challenges must be addressed for successful translation. Ensuring consistent production under GMP standards is critical to maintaining safety, efficacy, and reproducibility. To overcome this, implementing standardized protocols and rigorous quality control measures is essential. Additionally, patient-specific variability in treatment responses remains a major hurdle. Utilizing advanced techniques, such as single-cell RNA sequencing, could aid in understanding and mitigating these responses by enabling biomarker-based patient stratification and tailored therapeutic protocols. Finally, further preclinical studies and adaptive clinical trial designs will be necessary to optimize therapeutic outcomes across diverse patient populations. These strategies collectively aim to bridge the gap between preclinical findings and clinical application, ensuring the successful translation of PD-MSCsPRL-1 into viable therapies.
In conclusion, this study suggest that PD-MSCsPRL-1 show significant potential for treating chronic liver diseases, particularly cirrhosis, by reducing fibrosis and promoting regeneration. These cells address current therapy limitations, offering enhanced outcomes through targeted modulation of TGFB1 and BMP7 pathways. This study highlights the promise of engineered stem cells in developing effective, pathway-specific therapies, positioning PRL-1-enhanced MSCs as a breakthrough in regenerative medicine.

FOOTNOTES

Authors’ contribution
Conceptualization: J.Y.K., S.H.B., and G.J.K.; Data curation: J.Y.K.; Formal analysis: J.Y.K.; Methodology: J.Y.K., H.P., S.Y.P., S.H.K., and J.Y.L; Investigation: J.Y.K. and G.J.K.; Validation: J.Y.K., K.S.L., and G.J.K.; Visualization: J.Y.K., H.P., and J.Y.L.; Resources: G.J.K.; Writing – original draft: J.Y.K. and S.H.K.; Writing – review & editing: J.Y.K., and G.J.K.; Supervision: G.J.K.; Project administration: G.J.K.; Funding acquisition: G.J.K.
Acknowledgements
This research was supported by the Basic Science Research Program through the National Research Foundation of Korea (NRF) (RS-2023-00279247) grant funded by the Korea Government (MSIT) and the Korean Fund for Regenerative Medicine (KFRM) grant funded by the Korea Government (RS-2022-00070304).
Conflicts of Interest
Gi Jin Kim is the owner of PLABiologics Co., Ltd. (hereafter referred to as PLABiologics) and holds stock in the company. The authors declare that this study has received research grants from the Korean Government. The Korean Government, as the funder, was not involved in any activities related to this study. The remaining authors have no conflicts of interest to declare.

SUPPLEMENTAL MATERIAL

Supplementary material is available at Clinical and Molecular Hepatology website (http://www.e-cmh.org).
Supplementary Table 1.
Primer sequences used for quantitative real-time polymerase chain reaction
cmh-2024-0741-Supplementary-Table-1.pdf
Supplementary Figure 1.
Hepatocyte proliferation by PRL-1 in rat hepatic epithelial cells. Representative IF images and quantification of Ki-67 (green) and HNF4A (red) in WB-F344 treated with TGFB1 (2 ng/mL) for 48 hours, followed by recombinant PRL-1 (5 pg/mL) for 24 hours and/or pentamidine (PRL-1 inhibitor, 1 μg/mL) for 30 minutes. Scale bar=100 μm. Values represent mean±standard deviation, as determined by one-way ANOVA. *P<0.05, **P<0.01.
cmh-2024-0741-Supplementary-Figure-1.pdf
Supplementary Figure 2.
PD-MSCsPRL-1 affects epithelial and mesenchymal phenotypes in rat hepatic epithelial cells. (A) Representative images in WB-F344 (a diploid epithelial cell line from normal adult rat liver with phenotypic properties of “oval” cells) treated with TGFB1 (2 ng/mL) for 48 and 72 hours and cocultured with PD-MSCs or PD-MSCsPRL-1. (B) mRNA expression of epithelial marker Cdh1, hepatocyte marker Alb, and mesenchymal markers (Vim and Snai1) in WB-F344. Scale bar=500 μm. Values represent mean±standard deviation, as determined by one-way ANOVA. ns, not significant. *P<0.05, **P<0.01, ***P<0.001.
cmh-2024-0741-Supplementary-Figure-2.pdf
Supplementary Figure 3.
Hepatic stellate cells (HSCs) do not reserve epithelial marker. (A) Representative images in T-HSC/Cl6 (a rat HSC line) treated with TGFB1 (2 ng/mL) for 48 and 72 hours and cocultured with PD-MSCs or PD-MSCsPRL-1. (B) mRNA expression of epithelial marker Cdh1, hepatocyte marker Alb, and mesenchymal markers (Vim and Snai1) in T-HSC/Cl6. Scale bar=500 μm. Values represent mean±standard deviation, as determined by Student’s t-test or one-way ANOVA. ns, not significant. *P<0.05, **P<0.01, ***P<0.001.
cmh-2024-0741-Supplementary-Figure-3.pdf
Supplementary Figure 4.
Overexpression of PRL-1 in PD-MSCs. (A) Exogenous PRL-1 in supernatant of PD-MSCs or PD-MSCsPRL-1 by ELISA. (B) Western blotting of BMP7 and PRL-1 in PD-MSCs or PD-MSCsPRL-1 in the presence or absence of siRNA-BMP7 (siBMP7; 50 nM) for 24 hours, normalized to GAPDH. (C) qPCR analysis of PRL-1 in PD-MSCs in response to 5, 50, and 500 pg/mL for 24 and 72 hours. Values represent mean±standard deviation, as determined by Student’s t-test or one-way ANOVA. **P<0.01, ***P<0.001.
cmh-2024-0741-Supplementary-Figure-4.pdf

Figure 1.
Placenta-derived mesenchymal stem cells engineered to overexpress phosphatase of regenerating liver-1 (PD-MSCsPRL-1) alleviates hepatic fibrosis in a bile duct ligation (BDL)-injured rat model. (A) Experimental scheme. (B) Histopathological analysis of rat liver sections stained with H&E and survival rate (right; n=5–21/group). (C) Histopathological analysis and quantifications of rat liver sections stained with Sirius red and Masson’s trichrome at 2 weeks post-transplantation of PD-MSCs and PD-MSCsPRL-1 (n=5/group). (D) Western blotting and quantifications of fibrosis markers (COL1A1, ACTA2) in rat livers (n=5–21/group), normalized to GAPDH. (E) Serum levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), and total bilirubin (n=14–21/group). (F) Serum Interleukin-(IL)-10 levels quantified by ELISA (n=14–21/group). Scale bar=100 μm. Value represents mean±standard deviation, as determined by one-way ANOVA. ns, not significant. *P<0.05, **P<0.01, ***P<0.001, ****P<0.0001.

cmh-2024-0741f1.jpg
Figure 2.
Placenta-derived mesenchymal stem cells engineered to overexpress phosphatase of regenerating liver-1 (PD-MSCsPRL-1) regulates cell survival and death in BDL rats. (A) Volcano plot showing upregulated and downregulated factors in the supernatant of PD-MSCs and PD-MSCsPRL-1. (B) Serum interleukin (IL)-6 levels quantified by ELISA (n=18–21/group). (C) Western blotting of liver regeneration markers (IL-6, GP130, t/p-STAT3, ALB) and correlation between IL-6 and p-STAT3 (n=4–5/group). Serum albumin levels (n=5–18/group). (D) Western blotting of antiapoptotic (BCL2) and proapoptotic (BAX, c-PARP, c-CASPASE-3) markers in bile duct ligation (BDL) rat liver protein. Quantification of c-CASPASE-3 (n=5–18/group). (E) Western blotting of cell survival markers (CDK4, CCND1) in BDL rat liver protein (n=5/group), normalized to GAPDH. (F) IHC images stained by PCNA and quantification of hepatocyte nuclei (strong: dark blue, moderate: regular blue, weak: light blue) (n=4–5/group). Scale bar=100 μm. Value represents mean±standard deviation, as determined by one-way ANOVA. ns, not significant. *P<0.05, **P<0.01, ****P<0.0001.

cmh-2024-0741f2.jpg
Figure 3.
Placenta-derived mesenchymal stem cells engineered to overexpress phosphatase of regenerating liver-1 (PD-MSCsPRL-1) downregulates the mesenchymal phenotype, repressing TGFB/SMAD signaling in fibrotic rat liver. (A) Serological TGFB1 and liver mRNA Tgfb1 expression in rats (n=4–5/group) and their correlation. (B) Histological mesenchymal morphology in hepatocytes (yellow dotted lines) of liver tissues. V, vein; H, hepatocytes; C, cholangiocytes. (C) qPCR of mesenchymal markers (Vim, Snai1) in BDL-injured rat liver at 1, 2, 3, and 5 weeks (n=4–5/group). (D) Western blotting of mesenchymal markers and p-SMAD2 in total and nuclear liver protein extracts (n=4–5/group), normalized to GAPDH for total protein and LMNB1 for nuclear protein. (E) IF of p-SMAD2 in injured rat livers at 2 weeks post-transplantation (arrows indicate merge, red=p-SMAD2, blue=DAPI). (F) Schematic of reversed EMT by repressing p-SMAD2, leading to decreased SNAI1 and VIM. Scale bar=100 μm. Data represent mean±standard deviation, analyzed by one-way ANOVA. EMT, epithelial-to-mesenchymal transition; IF, immunofluorescence; ns, not significant. *P<0.05, **P<0.01, ****P<0.0001.

cmh-2024-0741f3.jpg
Figure 4.
Placenta-derived mesenchymal stem cells engineered to overexpress phosphatase of regenerating liver-1 (PD-MSCsPRL-1) induces BMP7 expression via SMAD1/5 signaling. (A) Serological BMP7 and liver mRNA Bmp7 expression in rats (n=4–5/group) and their correlation. (B) Western blot analysis of BMP7 and t/p-SMAD1/5 expression in rat livers (n=4–5/group). (C) IHC of BMP7 and PRL-1 in hepatocytes of bile duct ligation (BDL)-induced liver tissues at 3 weeks post-transplantation with PD-MSCs or PD-MSCsPRL-1 (n=4–5/group). Correlation between BMP7 and PRL-1 intensities in IHC (n=4–5/group). (D) qPCR of epithelial markers (Dsp, Cdh1, Tjp1) in rat livers (n=4–5/group). (E) Western blotting of TJP and CDH1, and co-staining of CDH1 (green) and BMP7 (red) in rat liver sections 3 weeks post-transplantation (blue=DAPI). (F) Schematic of enhanced MET via increased p-SMAD1/5, leading to elevated TJP and CDH1 levels. Scale bar=100 μm. Data represent mean±standard deviation, analyzed by one-way ANOVA. ns, not significant. *P<0.05, **P<0.01, ***P<0.001, ****P<0.0001.

cmh-2024-0741f4.jpg
Figure 5.
Placenta-derived mesenchymal stem cells engineered to overexpress phosphatase of regenerating liver-1 (PD-MSCsPRL-1) coculture retains the epithelial phenotype of rat liver epithelial cells. (A) Schematic of PD-MSCs or PD-MSCsPRL-1 coculture with hepatocytes exposed to TGFB1 for 48 hours and siRNA-PRL-1 (siPRL-1; 50 nM) for 24 hours. Correlation of TGFB1 and BMP7 by ELISA. Western blotting of PRL-1 and ALB. IF showing CDH1 (green) and BMP7 (red) expression. Quantification of CDH1. (B) Western blotting of BMP7, CDH1, and t/p-SMAD1/5 in hepatocytes. (C) Exogenous BMP7 and TGFB1 in supernatant by ELISA. (D) qPCR of epithelial (Bmp7, Dsp, Cdh1) and mesenchymal (Tgfb1, Snai1, Col1a1) markers. (E) Western blotting of COL1A1, SNAI1, and t/p-SMAD2. (F) Schematic of TGFB1/BMP7 balance regulating EMT/MET. Scale bar=50 μm. Data represent mean±standard deviation, analyzed by one-way ANOVA. ns, not significant. *P<0.05, **P<0.01, ***P<0.001, ****P<0.0001.

cmh-2024-0741f5.jpg
Figure 6.
Phosphatase of regenerating liver-1 (PRL-1) regulates BMP7 expression. (A) Exogenous BMP7 level in the supernatant and western blotting for BMP7 and PRL-1 in the lysates of PD-MSCs or PD-MSCsPRL-1 in presence or absence of siPRL-1. BMP7 protein expression in placenta-derived mesenchymal stem cells (PD-MSCs) in presence of PRL-1 (5 pg/mL) for 24 hours. (B) Fluorescence image showing the co-localization of BMP7 (green) and PRL-1 (red) in WB-F344 cells (blue=DAPI). Western blotting for the interaction between BMP7 and PRL-1 using a Co-immunoprecipitation assay. (C) Western blotting of p-SMAD1/5 and p-SMAD2 in WB-F344 cells exposed to TGFB1 and cocultured with PD-MSCs or PD-MSCsPRL-1 with or without siRNA BMP7 (siBMP7; 50 nM) for 24 hours. (D) Wound healing assay and quantification in WB-F344 cells in response to TGFB1, followed by PRL-1 and/or pentamidine (PRL-1 inhibitor, 1 μg/mL) for 30 min. (E) Western blotting of p-SMAD1/5 and p-SMAD2 in WB-F344, normalized to t-SMAD1/5 and t-SMAD2, respectively. (F) Schematic of TGFB1/BMP7 balance by PRL-1 through SMAD1/2/5 in hepatocytes regulating epithelial-to-mesenchymal transition (EMT)/mesenchymal-to-epithelial transition (MET). Scale bar=50 μm. Data represent mean±standard deviation, analyzed by Student’s t-test or one-way ANOVA. *P<0.05, **P<0.01, ***P<0.001.

cmh-2024-0741f6.jpg

cmh-2024-0741f7.jpg

Abbreviations

BDL
bile duct ligation
CCl4
carbon tetrachloride
EMT
epithelial-to-mesenchymal transition
MET
mesenchymal-to-epithelial transition
PD-MSCs
placenta-derived mesenchymal stem cells
PRL-1
phosphatase of regenerating liver-1
SNAI1
snail family transcriptional repressor 1
STAT3
signal transducer and activator of transcription 3
TGFB1
transforming growth factor B1

REFERENCES

1. Kalluri R, Weinberg RA. The basics of epithelial-mesenchymal transition. J Clin Invest 2009;119:1420-1428.
crossref pmid pmc
2. Hay ED. An overview of epithelio-mesenchymal transformation. Acta Anat (Basel) 1995;154:8-20.
crossref pmid pdf
3. Oh SH, Swiderska-Syn M, Jewell ML, Premont RT, Diehl AM. Liver regeneration requires Yap1-TGFβ-dependent epithelial-mesenchymal transition in hepatocytes. J Hepatol 2018;69:359-367.
crossref pmid pmc
4. Zhao YL, Zhu RT, Sun YL. Epithelial-mesenchymal transition in liver fibrosis. Biomed Rep 2016;4:269-274.
crossref pmid pmc
5. Lee SJ, Kim KH, Park KK. Mechanisms of fibrogenesis in liver cirrhosis: The molecular aspects of epithelial-mesenchymal transition. World J Hepatol 2014;6:207-216.
crossref pmid pmc
6. Lin SZ, Chang YJ, Liu JW, Chang LF, Sun LY, Li YS, et al. Transplantation of human Wharton’s Jelly-derived stem cells alleviates chemically induced liver fibrosis in rats. Cell Transplant 2010;19:1451-1463.
crossref pmid pdf
7. Jung J, Choi JH, Lee Y, Park JW, Oh IH, Hwang SG, et al. Human placenta-derived mesenchymal stem cells promote hepatic regeneration in CCl4 -injured rat liver model via increased autophagic mechanism. Stem Cells 2013;31:1584-1596.
crossref pmid pdf
8. Lee YB, Choi JH, Kim EN, Seok J, Lee HJ, Yoon JH, et al. Human chorionic plate-derived mesenchymal stem cells restore hepatic lipid metabolism in a rat model of bile duct ligation. Stem Cells Int 2017;2017:5180579.
crossref pmid pmc pdf
9. Ocansey DKW, Pei B, Yan Y, Qian H, Zhang X, Xu W, et al. Improved therapeutics of modified mesenchymal stem cells: an update. J Transl Med 2020;18:42.
crossref pmid pmc pdf
10. Campbell AM, Zhang ZY. Phosphatase of regenerating liver: a novel target for cancer therapy. Expert Opin Ther Targets 2014;18:555-569.
crossref pmid pmc
11. Liu LZ, He YZ, Dong PP, Ma LJ, Wang ZC, Liu XY, et al. Protein tyrosine phosphatase PTP4A1 promotes proliferation and epithelial-mesenchymal transition in intrahepatic cholangiocarcinoma via the PI3K/AKT pathway. Oncotarget 2016;7:75210-75220.
crossref pmid pmc
12. Kim JY, Choi JH, Jun JH, Park S, Jung J, Bae SH, et al. Enhanced PRL-1 expression in placenta-derived mesenchymal stem cells accelerates hepatic function via mitochondrial dynamics in a cirrhotic rat model. Stem Cell Res Ther 2020;11:512.
crossref pmid pmc pdf
13. Lee MJ, Jung J, Na KH, Moon JS, Lee HJ, Kim JH, et al. Antifibrotic effect of chorionic plate-derived mesenchymal stem cells isolated from human placenta in a rat model of CCl(4)-injured liver: potential application to the treatment of hepatic diseases. J Cell Biochem 2010;111:1453-1463.
crossref pmid
14. Kim JY, Choi JH, Kim SH, Park H, Lee D, Kim GJ. Efficacy of gene modification in placenta-derived mesenchymal stem cells based on nonviral electroporation. Int J Stem Cells 2021;14:112-118.
crossref pmid pmc
15. Kountouras J, Billing BH, Scheuer PJ. Prolonged bile duct obstruction: a new experimental model for cirrhosis in the rat. Br J Exp Pathol 1984;65:305-311.
pmid pmc
16. Kim JY, Jun JH, Park SY, Yang SW, Bae SH, Kim GJ. Dynamic regulation of miRNA expression by functionally enhanced placental mesenchymal stem cells promoteshepatic regeneration in a rat model with bile duct ligation. Int J Mol Sci 2019;20.
crossref pmid
17. Xiang X, Feng D, Hwang S, Ren T, Wang X, Trojnar E, et al. Interleukin-22 ameliorates acute-on-chronic liver failure by reprogramming impaired regeneration pathways in mice. J Hepatol 2020;72:736-745.
crossref pmid pmc
18. Wells RG. The epithelial-to-mesenchymal transition in liver fibrosis: here today, gone tomorrow? Hepatology 2010;51:737-740.
crossref pmid pmc
19. Zeisberg M, Hanai J, Sugimoto H, Mammoto T, Charytan D, Strutz F, et al. BMP-7 counteracts TGF-beta1-induced epithelial-to-mesenchymal transition and reverses chronic renal injury. Nat Med 2003;9:964-968.
crossref pmid pdf
20. Shi Y, Hu G, Su J, Li W, Chen Q, Shou P, et al. Mesenchymal stem cells: a new strategy for immunosuppression and tissue repair. Cell Res 2010;20:510-518.
crossref pmid pdf
21. Lee YA, Wallace MC, Friedman SL. Pathobiology of liver fibrosis: a translational success story. Gut 2015;64:830-841.
crossref pmid pmc
22. Hao Y, Baker D, Ten Dijke P. TGF-β-mediated epithelialmesenchymal transition and cancer metastasis. Int J Mol Sci 2019;20.
crossref pmid
23. Li T, Yan Y, Wang B, Qian H, Zhang X, Shen L, et al. Exosomes derived from human umbilical cord mesenchymal stem cells alleviate liver fibrosis. Stem Cells Dev 2013;22:845-854.
crossref pmid pmc
24. Wang SL, Yang CQ, Qi XL, Yuan M, Chang YZ, Yang L, et al. Inhibitory effect of bone morphogenetic protein-7 on hepatic fibrosis in rats. Int J Clin Exp Pathol 2013;6:897-903.
pmid pmc
25. Cervantes-Garcia D, Cuellar-Juarez AG, Borrego-Soto G, Rojas-Martinez A, Aldaba-Muruato LR, Salinas E, et al. Adenoviral‑bone morphogenetic protein‑7 and/or doxazosin therapies promote the reversion of fibrosis/cirrhosis in a cirrhotic hamster model. Mol Med Rep 2017;16:9431-9440.
crossref pmid pmc
26. Dituri F, Cossu C, Mancarella S, Giannelli G. The interactivity between TGFβ and BMP signaling in organogenesis, fibrosis, and cancer. Cells 2019;8.
crossref pmid
27. Chung YH, Huang YH, Chu TH, Chen CL, Lin PR, Huang SC, et al. BMP-2 restoration aids in recovery from liver fibrosis by attenuating TGF-β1 signaling. Lab Invest 2018;98:999-1013.
crossref pmid pdf
28. Zhong L, Wang X, Wang S, Yang L, Gao H, Yang C. The antifibrotic effect of bone morphogenic protein-7(BMP-7) on liver fibrosis. Int J Med Sci 2013;10:441-450.
crossref pmid pmc
29. Islam SS, Mokhtari RB, Kumar S, Maalouf J, Arab S, Yeger H, et al. Spatio-temporal distribution of Smads and role of Smads/TGF-β/BMP-4 in the regulation of mouse bladder organogenesis. PLoS One 2013;8:e61340.
crossref pmid pmc
30. Sugimoto H, Yang C, LeBleu VS, Soubasakos MA, Giraldo M, Zeisberg M, et al. BMP-7 functions as a novel hormone to facilitate liver regeneration. Faseb j 2007;21:256-264.
crossref pmid pdf
31. Zeng Q, Hong W, Tan YH. Mouse PRL-2 and PRL-3, two potentially prenylated protein tyrosine phosphatases homologous to PRL-1. Biochem Biophys Res Commun 1998;244:421-427.
crossref pmid

Editorial Office
The Korean Association for the Study of the Liver
Room A1210, 53 Mapo-daero(MapoTrapalace, Dowha-dong), Mapo-gu, Seoul, 04158, Korea
TEL: +82-2-703-0051   FAX: +82-2-703-0071    E-mail: cmh_journal@ijpnc.com
Copyright © The Korean Association for the Study of the Liver.         
COUNTER
TODAY : 2821
TOTAL : 2897323
Close layer