Skip to main content
PLOS One logoLink to PLOS One
. 2025 Apr 25;20(4):e0321020. doi: 10.1371/journal.pone.0321020

Solute carrier family 2 member 2 (glucose transporter 2): a common factor of hepatocyte and hepatocellular carcinoma differentiation

Yejin Kim 1,#, Yu Yeuni 2,#, Hye Jin Heo 3,#, Eun Sun Kim 4, Kyungjae Myung 4, Ninib Baryawno 5, Yun Hak Kim 3,6,*, Chang-Kyu Oh 1,7,*
Editor: Heping Cao8
PMCID: PMC12026939  PMID: 40279337

Abstract

GLUT2 (SLC2A2), a vital glucose transporter in liver, pancreas, and kidney tissues, regulates blood glucose levels and energy metabolism. Beyond its metabolic role, SLC2A2 contributes to cell differentiation and metabolic adaptation during embryogenesis and tissue regeneration. Despite its significance, the role of SLC2A2 in liver differentiation and hepatocellular carcinoma (HCC) remains underexplored. This study investigated SLC2A2’s role in liver differentiation using in silico, in vitro, and in vivo approaches. Analysis of GEO datasets (GSE132606, GSE25417, GSE67848) and TCGA HCC data revealed that while SLC2A2 expression decreases with HCC progression, stemness-associated genes, including SOX2 and POU5F1, are upregulated. Zebrafish embryos injected with SLC2A2-targeting morpholino exhibited reduced expression of the liver differentiation marker fabp10a without significantly altering the hepatoblast marker hhex. In HepG2 cells, SLC2A2 knockdown increased stemness and IGF1R pathway markers, indicating a shift toward less differentiated states. These findings suggest that SLC2A2 supports liver differentiation by regulating glucose metabolism and suppressing pathways associated with stemness and malignancy. Targeting SLC2A2 may serve as a promising therapeutic strategy for liver-related diseases, particularly HCC, by addressing its dual role in differentiation and tumor progression. Further mechanistic studies are warranted to fully elucidate these processes.

Introduction

Hepatocellular carcinoma (HCC) remains a significant global health challenge due to its high prevalence, poor prognosis, and the limited efficacy of current therapeutic options [1,2]. While traditional treatments such as surgery, chemotherapy, and targeted therapies have been implemented, their effectiveness is often compromised by the molecular heterogeneity of HCC and its aggressive nature [3,4]. Consequently, there is growing interest in the molecular classification of HCC to more effectively tailor treatments by targeting specific genetic alterations [5,6]. Recent advancements in this area emphasize the critical role of molecular classification, which categorizes tumors based on distinct genetic and molecular profiles, thereby identifying key genes and pathways as potential therapeutic targets [7,8]. This strategy paves the way for more precise and personalized treatment modalities [9]. Nevertheless, the prognosis for HCC patients remains dire, highlighting the necessity for ongoing research into novel therapeutic targets [10].

One promising direction in HCC research involves the study of oncofetal genes, which are typically expressed during fetal development and re-expressed in various cancers, including HCC [11,12]. These genes play pivotal roles in cancer progression due to their involvement in cell proliferation and vascularization—processes that are crucial both in embryonic development and tumor growth [13]. This similarity has led to the concept of “oncofetal reprogramming,” which seeks to understand and exploit the parallels between embryonic development and cancer progression for therapeutic gain [14,15]. Recent studies have shown that reprogramming of oncofetal genes in HCC plays a crucial role in promoting tumor progression and immune evasion [16]. This research highlights the significance of targeting these genes within the tumor microenvironment, suggesting that such an approach could offer a more precise and effective treatment strategy compared to traditional therapies [17,18]. This approach differentiates itself by directly addressing the underlying mechanisms driven by oncofetal genes, offering new potential in the fight against HCC [19,20].

In the context of oncofetal research, the zebrafish (Danio rerio) model has emerged as a powerful tool for in vivo studies of liver development and cancer [21]. Zebrafish embryos provide a unique platform for investigating the role of specific genes in liver differentiation and tumorigenesis, facilitating a comprehensive integration of in silico, in vitro, and in vivo methodologies [22]. Recent studies have highlighted the critical role of key transcription factors regulated by signaling molecules in liver development, with zebrafish serving as an ideal model system to unravel these complex interactions [23,24]. Additionally, the zebrafish model has been successfully used to replicate liver tumorigenesis driven by specific oncogenes, offering valuable insights into the molecular pathways involved in hepatocellular carcinoma [25,26]. These findings suggest the versatility and effectiveness of zebrafish as a model organism in advancing our understanding of liver biology and cancer [27].

Solute carrier family 2 member 2 (SLC2A2) is known to play a role in glucose uptake and metabolic regulation in various tissues, including pancreatic beta cells and renal tubular cells, thereby supporting cell growth and energy homeostasis. Its ability to regulate intracellular glucose levels is critical for maintaining cellular functions and promoting anabolic processes, particularly in rapidly growing or differentiating cells. Additionally, SLC2A2 plays a crucial role during embryogenesis, aiding in cell differentiation and metabolic adaptation at early developmental stages. This SLC2A2 has also been reported to be involved in the differentiation of liver and HCC progression [28]. Our study aims to identify novel therapeutic targets that can improve the prognosis for HCC patients by investigating the expression and functional dynamics of SLC2A2 in correlation with liver differentiation markers [29,30]. Through a detailed exploration of SLC2A2 role in these processes, using a combination of molecular classification techniques and the zebrafish model [31], this research will enhance our understanding of the molecular mechanisms driving HCC progression and contribute to the development of targeted therapies that overcome the limitations of current treatment paradigms [5,32].

Materials and methods

Data collection

Three datasets (GSE132606 [33], GSE25417 [34], and GSE67848 [35]) were obtained from the Gene Expression Omnibus database to investigate the relationship between SLC2A2 expression and stemness over time. Each dataset contained gene expression profiles measured at multiple time points. The fragments per kilobase of transcript per million mapped for liver cancer RNA-sequencing data from The Cancer Genomic Atlas (TCGA) was downloaded from the Genomic Data Commons, and patients with unavailable clinical information were excluded.

Statistical and bioinformatic analysis

Statistical analyses were performed to assess the significance of the relationship between SLC2A2 expression and stemness. Correlation analysis and Spearman’s correlation coefficients were used to quantify the strength and direction of the association between SLC2A2 and stemness gene expression levels. Additionally, statistical tests such as analysis of variance or t-tests were conducted to compare the expression levels of SLC2A2 and stemness markers at different time points or experimental conditions. In R Programming Language software, the R package “ggscatter” was used to correlation analysis.

Zebrafish maintenance

Wild-type AB zebrafish were obtained from the Korea Zebrafish Core Resource Center (KZRC) and maintained in an automatic circulation system (Genomic-Design) at 28.5°C. All experiments involving zebrafish were cRonducted in accordance with the Institutional Animal Care and Use Committee (IACUC) guidelines of Pusan National University (PNU-2023–0359). Zebrafish embryos used in the experiments were cultured in E3 media (5 mM NaCl, 0.17 mM KCl, 0.4 mM CaCl2, and 0.16 mM MgSO4) in incubators set at 28°C. Zebrafish embryos were sacrificed under deep anesthesia using tricaine at a concentration of 6 g/L, which exceeds the standard anesthetic dose (2 g/L) to ensure rapid and humane euthanasia. This method was chosen to minimize potential suffering.

Morpholino injection

A splice-blocking morpholino targeting exon3/intron3 of SLC2A2 (Gene Tools) was dissolved in DEPC water at 25 ng/nL stock. The sequence of morpholino targeting SLC2A2 is 5′-CAAGTTCACAGATACTCCACCTTCC-3′. A morpholino targeting SLC2A2 was injected into embryos of wild-type AB zebrafish at the one-cell stage post-fertilization. Microinjections were performed using a FemtoJet 4i microinjector (Eppendorf, Hamburg, Germany).

RNA isolation and quantitative PCR using zebrafish embryos

To isolate zebrafish embryos, they were homogenized using RNase-free pestles. Total RNA was extracted from the homogenized embryos using 1 mL TRIzol reagent (Molecular Research Center Inc.) manually. Chloroform was added to separate proteins, and isopropanol was added to precipitate total RNA. Using SuperScript IV Reverse Transcriptase (Thermo Fisher Scientific, Waltham, MA, USA) 3 μg of total RNA was reverse-transcribed. Real-time polymerase chain reaction (RT-PCR) was performed using GoTaq G2 DNA Polymerase (Promega), and the results were visualized using BANDi-Green Nucleic Acid Stain (Translab, Daejeon, Korea). A quantitative RT-PCR (qPCR) assay was performed using PowerUp SYBR Green Master Mix (Thermo Fisher Scientific). The sequence of primers used for RT-qPCR are listed in Table 1. We analyzed the target genes expression levels using the comparative threshold method, and the results were normalized to β-actin as endogenous control.

Table 1. Primers used for qPCR gene expression analysis.

Gene symbol Forward primer Reverse primer
AFP acatcctcagcttgctgtct aatgcttggctctcctggat
CK19 ctttgtgtcctcgtcctcct gtcgcggatcttcacctcta
DLK gcttcatcgacaagacctgc caggtctcgcacttgttgag
EPCAM cagaaggagatcacaacgcg tccagatccagttgttcccc
GLUT2 ccagaaagccccagatacctttac cagcatcagtgccactagaatagg
IGF1 catgtcctcctcgcatctcttcta atctccagcctccttagatcacag
IGF1R aaaccttcgcctcatcctaggaga tttatgtcccctttgctttggcgc
IGF2 caatggggaagtcgatgctg ggaaacagcactcctcaacg
Nanog tgagtgtggatccagcttgt tctctgcagaagtgggttgt
PROM1 ttcttgaccgactgagaccc ccaagcacagagggtcattg
SALL4 atttgtgggaccctcgacat ctgagttattgttcgccccg
Sox2 tgatggagacggagctgaag gcttgctgatctccgagttg
Sox9 atgaagatgaccgacgagca aacttgtcctcctcgctctc
GAPDH catgttcgtcatggggtgaacca agtgatggcatggactgtggtcat
g6pc1a.1 tcacagcgttgctttcaatc acttggtgtgggaaatgagc
Igf1a cacactgtccttccccaagt gatgaccagggcgtagttgt

Whole-mount in situ hybridization (WISH)

WISH was performed on 2- and 4-d post-fertilization (dpf) embryos. The embryos were fixed in 4% paraformaldehyde in phosphate-buffered saline and dehydrated with methanol at -20°C overnight. The samples were incubated with acetone at -20°C for permeabilization and were hybridized with a digoxigenin (DIG)-labeled antisense RNA probe in a hybridization buffer (50% formamide, 5× SSC, 500 μg/mL Torula yeast tRNA, 50 μg/mL heparin, 0.1% Tween-20, and 9 mM citric acid; pH 6.5) for 3 d. The samples were then washed using 2× and 0.2× SSC solutions. The washed samples were blocked with normal goat serum and bovine serum albumin (BSA) and then incubated with alkaline phosphatase-conjugated DIG antibodies (1:5000) (Roche, Basel, Switzerland) overnight at 4°C. The samples were finally incubated with alkaline phosphatase reaction buffer (100 mM Tris; pH 9.5), 50 mM MgCl2, 100 mM NaCl, and 0.1% Tween-20, and the NBT/BCIP substrate (Promega) was used to visualize the WISH signal.

Statistical analysis of zebrafish experiments

For statistical analysis of the zebrafish experiments, Student’s t-test was used, and all experiments were performed in triplicate. The figures and graphs represent averages of three independent experiments. The error bars indicate the standard error of the mean. p-values <0.05 were considered statistically significant.

Cell culture

Human HCC cells (HepG2) were purchased from the Korean Cell Line Bank. HepG2 cells were cultured in Dulbecco’s modified Eagle medium supplemented with 10% fetal bovine serum (Gibco, CA, USA) at 37°C with 5% CO2.

Gene expression by lentiviral infection

Lentiviral particles were generated for target gene expression and infection, as described in a previous study [36]. The target sequence was 5′-ACCAATTCCAGCTACCGAC-3′ (shSLC2A2), and 5′- CGAGATCTATGGACTACAAGGACGACGATGACAAGATGACAGA

AGATAAGGTCA-3′ (SLC2A2 over).

qPCR using cell line

Total RNA was extracted using a RNeasy Mini Kit (Qiagen, Hilden, Germany). Complementary DNA was synthesized using a Smart Gene Compact cDNA Synthesis Kit (Smart Gene, South Korea). qPCR was performed using the LightCycler 96 Real-Time PCR System (Roche). Target mRNA expression relative to the housekeeping gene expression (GAPDH) was calculated using the ΔΔCT method. The sequence of primers used for RT-qPCR are listed in Table 1.

Western blotting

HepG2 cells were harvested, homogenized in lysis buffer, and centrifuged at 13,000 rpm for 20 min at 4°C. In total, 30 μg of protein underwent 10% sodium dodecyl sulfate-polyacrylamide gel electrophoresis. Proteins were then transferred onto nitrocellulose membranes that were blocked with 5% BSA, and incubated with antibodies against anti-GLUT2 (ABclonal Technology, MA, USA) overnight at 4°C (1:500 dilution). The membranes were then probed with an anti-β-actin antibody (ABclonal Technology) for 3 h (1:1,000 dilution) as an internal control. Antigen-antibody complexes were detected using chemiluminescence (Thermo Fisher Scientific).

Fluorescent Imaging of Transgenic Zebrafish Embryos

Fabp10a:mCherry transgenic zebrafish embryos were used for fluorescent imaging. Embryos from the control group and those injected with slc2a2-MO were imaged at 72, 96, and 120 hours post-fertilization (hpf). Prior to imaging, embryos were anesthetized using tricaine (0.016% w/v) and mounted in 3% methylcellulose to ensure stability during imaging. Fluorescent signals were captured using a fluorescent stereo microscope (Zeiss, Discovery V8)dissecting fluorescence microscope equipped with appropriate filters for detecting mCherry fluorescence. Images were acquired under identical conditions for all experimental groups to allow for consistent comparison.

Immunohistochemistry using zebrafish embryos

Zebrafish embryos were fixed in 4% paraformaldehyde at 4°C overnight, washed twice with PBT (PBS + 0.1% Tween-20), dehydrated through a PBT/MeOH gradient, and incubated in 100% MeOH at -20°C overnight. After rehydration through a MeOH/PBT gradient, samples were incubated in 1% Triton X-100 for 1 hour at room temperature, washed twice with PBT, and blocked for 1 hour at room temperature. Primary antibody incubation was performed with IGF1R antibody (A0243, ABclonal) diluted 1:100 in antibody buffer for 1–3 days at 4°C. Samples were then washed with sodium phosphate buffer and incubated with a 1:100 dilution of secondary antibody in the dark for 1 hour at room temperature. After washing with PBT, samples were imaged using a fluorescent stereo microscope (Zeiss, Discovery V8)

Statistical analysis

All data were analyzed using Student’s t-test, and p-values were used to assess statistical significance as follows: * p <0.05, ** p <0.01, and *** p <0.001.

Results

Negative correlation between SLC2A2 and liver stemness-related genes

We investigated temporal changes in the expression profiles of mature liver markers in the GSE132060, GSE25417, and GSE67848 datasets to validate the data. The expression patterns of albumin, G6PC, and CYP3A4 showed an upward trend in these datasets (Fig 1A). Conversely, we observed a simultaneous decrease in the expression levels of stemness-associated genes NANOG, SOX2, and POU5F1 during liver development (Fig 1B). The GSE132060, GSE25417, and GSE67848 datasets also demonstrated a consistent increase in SLC2A2 expression during development across the three datasets (Fig 1C).

Fig 1. Gene expression dynamics during liver development.

Fig 1

(A) Gene expression pattern of albumin, G6PC, and CYP3A4 in GSE132060, GSE25417, and GSE67848 datasets. (B) Gene expression pattern of NANOG, SOX2, and POU5F1 in GSE132060, GSE25417, and GSE67848 datasets. (C) Gene expression pattern of SLC2A2 in GSE132060, GSE25417, and GSE67848 datasets.

Negative correlation between SLC2A2 and stemness-related genes

To delineate SLC2A2 attributes in HCC, we investigated its expression dynamics across different cancer stages using the TCGA HCC dataset. Our analysis revealed a consistent decrease in SLC2A2 expression with increasing cancer stage. Conversely, SOX2 and POU5F1 expression levels progressively increased with advancing cancer stage (Fig 2A). A significant inverse correlation was observed between SLC2A2 and both stemness-associated (SOX2 and POU5F1) and oncofetal marker genes (AFP, SALL4, FOXM1, and IGF2BP1) (Fig 2B2C).

Fig 2. Changes in SLC2A2 gene expression in hepatocellular carcinoma (HCC) based on its stage and correlation with oncofetal and stemness genes.

Fig 2

(A) The bar plot illustrates the expression levels of SLC2A2, SOX2, and POU5F1 genes in HCC samples at different stages (Stages I, II, and III). (B) The scatter plot depicts the correlation between the expression levels of the SLC2A2, stemness (SOX2 and POU5F1), and (C) oncofetal genes (AFP, SALL4, FOXM1, and IGF2BP1). Each data point represents an individual sample, and the correlation coefficients and p-values are indicated.

Knockdown of SLC2A2 (GLUT2) in HepG2 cells increases the expression of oncofetal and differentiation-related genes

To investigate SLC2A2 function, SLC2A2 was knocked down in HepG2. SLC2A2 knockdown was validated using qRT-PCR (Fig 3A) and western blotting (Fig 3B and S1 Fig). Subsequently, stemness markers were examined after SLC2A2 genetic regulation in HepG2. Stem cell markers expression, including AFP, CK19, DLK1, EPCAM, IGF2, PROM1, and SALL4, increased after SLC2A2 knockdown. In contrast, SLC2A2 overexpression reduced PROM1, CK19, DLK1, SALL4 and EPCAM expression (Fig 3C).

Fig 3. SLC2A2 is associated with IGF1R pathways differentiation.

Fig 3

(A) SLC2A2 expression levels were determined using quantitative real-time polymerase chain reaction (qPCR) in or GLUT2 overexpression cells compared with control cells (n = 4). (B) GLUT2 protein levels were measured by western blotting (n = 3). (C) We confirmed whether the difference in SLC2A2 expression level was related to stemness (n = 3) or (D) the IGF1R pathways (n = 3) via qPCR. * p <0.05, ** p <0.01, and *** p <0.001.

slc2a2 is essential for liver differentiation during vertebrate development

To optimize the morpholino dosage, a series of concentrations were injected into the embryos. Among the different doses tested, 5 ng/embryo was ideal for achieving sufficient slc2a2 knockdown (Fig 4A). The embryo phenotype was also observed after injecting different doses of morpholino. At a higher dose of 10 ng/embryo, off-target effects such as reduced brain and eyeball size and heart edema were observed at 5 dpf (Fig 4B). Therefore, a 5 ng/embryo dose was chosen as it achieved effective knockdown without inducing these off-target phenotypes.

Fig 4. SLC2A2 is essential for liver differentiation in developing vertebrates.

Fig 4

(A) RT‐PCR analysis of SLC2A2 and β‐actin using 4 dpf zebrafish embryos. Total RNA was isolated from uninjected control and SLC2A2 MO‐injected embryos (2.5, 5, and 10 ng). (B) Lateral view of zebrafish embryos after SLC2A2 MO-injection (2.5, 5, and 10 ng). The black arrow indicates heart edema in zebrafish embryos injected with 10 ng of SLC2A2-MO. (C) WISH images of uninjected embryos and SLC2A2-targeting morpholino-injected embryos at 5 dpf using fabp10a. The yellow line indicates fabp10a signal at the liver. (D) Using hepatoblast marker hhex, we captured WISH images of uninjected embryos and SLC2A2-targeting morpholino-injected embryos at 2 dpf. The white arrow indicates the hhex signal in the hepatoblast. (E) qRT-PCR analysis of fabp10a and hhex expression in uninjected embryos and SLC2A2-targeting morpholino-injected embryos at 4 dpf. mRNA expression is normalized to that of β-actin mRNA levels (*** indicates significance at p-value <0.001). Scale bars indicates 200 um. (F) Quantitative RT-PCR was used to measure hhex expression, normalized to β-actin mRNA levels. There was no significant difference (n.s.) observed in hhex expression between control and SLC2A2 MO-injected embryos. Data are represented as mean ± standard error of the mean (SEM). (G) Quantitative RT-PCR analysis was performed to evaluate the expression levels of igf1r. The mRNA expression levels were normalized to β-actin. A significant increase (p-value < 0.001, ***) in igf1r expression was observed in SLC2A2 MO-injected embryos compared to the control group. Data are presented as mean ± standard error of the mean (SEM). (G) Reduction in liver fluorescence intensity in zebrafish embryos following slc2a2 morpholino (MO) injection. Representative fluorescence microscopy images and quantitative analysis of liver fluorescence intensity in zebrafish embryos at 72, 96, and 120 hours post-fertilization (hpf). Uninjected embryos show consistent fluorescence in the liver across all time points, while embryos injected with slc2a2 MO exhibit a progressive reduction in fluorescence intensity. The fluorescence signal in slc2a2 MO-injected embryos decreased by approximately 60% at 96 hpf and 80% at 120 hpf compared to the uninjected controls.

Subsequently, the expression of liver differentiation markers was examined using whole mount in situ hybridization (WISH) and qPCR. Knock-down of slc2a2 resulted in the reduced expression of fabp10a in the liver at 5 dpf, as observed by WISH (Fig 4C). Additionally, slc2a2 knockdown decreased the expression of mature hepatocyte markers, including fabp10a and g6pc1a.1, as shown by qPCR (Fig 4E). However, slc2a2 knockdown did not significantly affect the expression of the hepatoblast marker hhex at 2 dpf, as assessed by WISH (Fig 4D) and qPCR (Fig 4F). Representative fluorescence microscopy images and quantitative analysis of liver fluorescence intensity in zebrafish embryos injected with slc2a2-MO compared to uninjected controls at 72, 96, and 120 hpf (Fig 4G).

IGF1R pathway is upregulated following slc2a2 knockdown in HepG2 cells and zebrafish embryos

Knockdown of slc2a2 using shGLUT2 significantly increased the expression of IGF1R and IGF2 compared to the pLKO control group. In contrast, overexpression of slc2a2 (GLUT2 over) significantly decreased IGF1R and IGF2 expression compared to the pMSCV control group. Notably, the reduction in IGF2 expression was confirmed through in vitro qPCR analysis (Fig 5A). RT-qPCR analysis further revealed a significant increase in IGF1R mRNA expression in zebrafish embryos following slc2a2 knockdown (Fig 5B). Additionally, protein level of IGF1R was measured in zebrafish embryos at 2 dpf via immunohistochemistry (IHC). The fluorescence intensity of IGF1R was markedly stronger in the slc2a2-MO group compared to the control group, indicating an increase in IGF1R protein expression (Fig 5C).

Fig 5. The IGF1R pathway is upregulated following slc2a2 knockdown in HepG2 cells and zebrafish embryos.

Fig 5

(A) qPCR analysis of IGF1R and IGF2 gene expression in HepG2 cells. Knockdown of slc2a2 using shGLUT2 significantly increased IGF1R and IGF2 expression compared to the pLKO control group (p < 0.05). Overexpression of slc2a2 (GLUT2 over) significantly decreased IGF1R and IGF2 expression compared to the pMSCV control group (p < 0.01 for IGF1R, p < 0.05 for IGF2). (B) qPCR analysis of igf1r mRNA expression in zebrafish embryos at 2 dpf. slc2a2 knockdown significantly increased igf1r expression compared to the control group (p < 0.001). (C) Immunohistochemistry (IHC) staining for IGF1R protein in zebrafish embryos at 2 dpf. slc2a2-MO treated embryos showed stronger IGF1R fluorescence intensity compared to control embryos, indicating increased IGF1R protein expression. All analyzed embryos (11/11) showed consistent results in both groups.

Discussion

SLC2A2 (GLUT2), a critical glucose transporter, plays a pivotal role in hepatic glucose metabolism but remains understudied in the context of liver differentiation [37]. This study integrated in silico, in vitro, and in vivo approaches to elucidate the relationship between SLC2A2 expression, liver differentiation, and its broader impact on progression of HCC.

Our analysis demonstrated that SLC2A2 expression was significantly reduced in HCC compared to other SLC2A family members (Fig 2A). This reduction correlated with increased expression of stem cell markers, such as PROM1, CK19, DLK1, SALL4, and EPCAM (Fig 3C), suggesting that low SLC2A2 levels may promote cancer stem cell traits, malignancy, and therapeutic resistance. These findings align with previous studies showing that diminished SLC2A2 expression correlates with poorer HCC prognosis [28].

In addition, our in vitro experiments revealed that SLC2A2 knockdown led to upregulation of IGF1R and IGF2 (Fig 5), key regulators of cell proliferation and survival [38]. This suggests that SLC2A2 may influence liver differentiation by modulating IGF1R signaling pathways. Increased IGF1R expressions likely suppresses differentiation by promoting cellular proliferation, a mechanism commonly observed in cancer biology [39]. This metabolic adaptation may be a compensatory response to glucose scarcity caused by reduced SLC2A2 expression, as similar mechanisms have been reported in other systems using C. elegans under starvation stress [38]. Targeting IGF1R in the context of SLC2A2 dysregulation presents a potential therapeutic approach for liver-related diseases, particularly HCC.

Our zebrafish model experiments provided further insights into the role of SLC2A2 in liver development. Morpholino-mediated knockdown of slc2a2 reduced the expression of fabp10a, a marker of mature hepatocytes, without significantly affecting the expression of hhex, a hepatoblast marker (Fig 4C4G). This indicates that while early liver development can proceed independently of SLC2A2, its role becomes increasingly critical as hepatoblasts differentiate into mature hepatocytes. The observed decrease in liver fluorescence intensity and increased IGF1R expression in slc2a2-knockdown embryos emphasize its regulatory function in liver differentiation (Fig 5C). The use of zebrafish as a model system is advantageous due to their rapid development, optical transparency, and genetic tractability, which allow for real-time observation of liver differentiation and gene-specific functional studies [40]. These features make zebrafish an ideal platform for investigating complex developmental and disease mechanisms in vivo.

By demonstrating parallels between fetal liver development and HCC progression, this study highlights the utility of zebrafish models in exploring the dual role of SLC2A2 in development and oncogenesis. The re-expression of fetal-like markers in HCC further supports the concept of oncofetal reprogramming, where developmental pathways are hijacked during cancer progression. These findings suggest the therapeutic potential of targeting SLC2A2 to modulate cancer stem cell traits and improve differentiation in HCC. However, further research is required to clarify the molecular mechanisms through which SLC2A2 regulates IGF1R signaling, particularly in the context of liver differentiation and HCC progression. Investigating how SLC2A2-mediated changes in glucose metabolism influence IGF1R pathway activation and downstream effects on cellular proliferation and differentiation will provide deeper insights into its role in liver biology and tumorigenesis.

In conclusion, our study demonstrates that SLC2A2 serves as a critical regulator of liver differentiation and HCC progression. The findings suggest that therapeutic strategies aimed at restoring SLC2A2 expression or counteracting its downstream effects may improve HCC prognosis and provide new avenues for liver-related disease treatments. Future studies should focus on unraveling the precise molecular mechanisms of SLC2A2 in hepatocyte differentiation and its potential as a therapeutic target in liver cancer.

Conclusions

This study demonstrates that SLC2A2 (GLUT2) plays a critical role in liver differentiation and HCC progression. In silico, in vitro, and in vivo analyses revealed that increased SLC2A2 expression promote hepatocyte differentiation, while its reduction correlates with advanced HCC stages and elevated stem cell markers. These findings highlight SLC2A2 as a potential therapeutic target for improving liver differentiation and suppressing cancer stem cell traits in HCC. Further research is needed to fully elucidate its mechanisms and translate these insights into clinical applications.

Supporting information

S1 Fig. Protein levels of GLUT2 were analyzed by western blotting.

GLUT2 protein expression was quantified compared to the control cells., ** p < 0.01, and *** p < 0.001.

(PPTX)

pone.0321020.s001.pptx (1.2MB, pptx)

Acknowledgments

None

Abbreviations

IGF1R

insulin-like growth factor 1 receptor

IGF2

insulin-like growth factor 2

SLC2A2

solute carrier family 2 member 2

GLUT

glucose transporter

HCC

hepatocellular carcinoma

TCGA

The Cancer Genomic Atlas

WISH

whole-mount in situ hybridization

DIG

digoxigenin

qPCR

quantitative real-time polymerase chain reaction

dpf

days post-fertilization

RT-PCR

real-time polymerase chain reaction

BSA

bovine serum albumin

HepG2

human HCC cells.

Data Availability

All GSE data files are available from the GEO database (GSE132606, GSE25417, GSE67848).

Funding Statement

This work was supported by the Bio & Medical Technology Development Program of the National Research Foundation (NRF) funded by the Korean government (MSIT) (No. RS-2023-00223764 to CO, RS-2023-00207946 to YHK).

References

  • 1.Gosalia AJ, Martin P, Jones PD. Advances and future directions in the treatment of hepatocellular carcinoma. Gastroenterol Hepatol (N Y). 2017;13(7):398–410. [PMC free article] [PubMed] [Google Scholar]
  • 2.Medavaram S, Zhang Y. Emerging therapies in advanced hepatocellular carcinoma. Exp Hematol Oncol. 2018;7:17. doi: 10.1186/s40164-018-0109-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Yang X, Yang C, Zhang S, Geng H, Zhu AX, Bernards R, et al. Precision treatment in advanced hepatocellular carcinoma. Cancer Cell. 2024;42(2):180–97. doi: 10.1016/j.ccell.2024.01.007 [DOI] [PubMed] [Google Scholar]
  • 4.Qiang Z, Wan J, Chen X, Wang H. Mechanisms and therapeutic targets of ErbB family receptors in hepatocellular carcinoma: a narrative review. Transl Cancer Res. 2024;13(6):3156–78. doi: 10.21037/tcr-24-837 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Alqahtani A, Author2 B, Author3 C. Hepatocellular carcinoma: molecular mechanisms and targeted therapies. Medicina (Kaunas). 2019;55(9):1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Critelli RM, De Maria N, Villa E. Biology of hepatocellular carcinoma. Dig Dis. 2015;33(5):635–41. [DOI] [PubMed] [Google Scholar]
  • 7.Coffin P, He A. Hepatocellular carcinoma: past and present challenges and progress in molecular classification and precision oncology. Int J Mol Sci. 2023;24(17). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Rebouissou S, Nault J-C. Advances in molecular classification and precision oncology in hepatocellular carcinoma. J Hepatol. 2020;72(2):215–29. doi: 10.1016/j.jhep.2019.08.017 [DOI] [PubMed] [Google Scholar]
  • 9.Teufel A. Current trends and advancements in the management of hepatocellular carcinoma. Dig Dis. 2024;42(4):349–60. [DOI] [PubMed] [Google Scholar]
  • 10.Gutierrez-Chakraborty E, et al. Discovering novel prognostic biomarkers of hepatocellular carcinoma using eXplainable artificial intelligence. bioRxiv. 2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Zhu Y, Xiao B, Liu M, Chen M, Xia N, Guo H, et al. N6-methyladenosine-modified oncofetal lncRNA MIR4435-2HG contributed to stemness features of hepatocellular carcinoma cells by regulating rRNA 2’-O methylation. Cell Mol Biol Lett. 2023;28(1):89. doi: 10.1186/s11658-023-00493-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Jeon A-J, Anene-Nzelu CG, Teo Y-Y, Chong SL, Sekar K, Wu L, et al. A genomic enhancer signature associates with hepatocellular carcinoma prognosis. JHEP Rep. 2023;5(6):100715. doi: 10.1016/j.jhepr.2023.100715 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Muller S. The oncofetal RNA-binding protein IGF2BP1 is a druggable, post-transcriptional super-enhancer of E2F-driven gene expression in cancer. Nucleic Acids Research. 2020;48(15):8576–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Cao J, Zhang Z, Zhou L, Luo M, Li L, Li B, et al. Oncofetal reprogramming in tumor development and progression: novel insights into cancer therapy. MedComm (2020). 2023;4(6):e427. doi: 10.1002/mco2.427 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Sharma A, Blériot C, Currenti J, Ginhoux F. Oncofetal reprogramming in tumour development and progression. Nat Rev Cancer. 2022;22(10):593–602. doi: 10.1038/s41568-022-00497-8 [DOI] [PubMed] [Google Scholar]
  • 16.Liu Y, Xun Z, Ma K, Liang S, Li X, Zhou S, et al. Identification of a tumour immune barrier in the HCC microenvironment that determines the efficacy of immunotherapy. J Hepatol. 2023;78(4):770–82. doi: 10.1016/j.jhep.2023.01.011 [DOI] [PubMed] [Google Scholar]
  • 17.Sharma A, Seow JJW, Dutertre C-A, Pai R, Blériot C, Mishra A, et al. Onco-fetal reprogramming of endothelial cells drives immunosuppressive macrophages in hepatocellular carcinoma. Cell. 2020;183(2):377-394.e21. doi: 10.1016/j.cell.2020.08.040 [DOI] [PubMed] [Google Scholar]
  • 18.Dzobo K. Taking a full snapshot of cancer biology: deciphering the tumor microenvironment for effective cancer therapy in the oncology clinic. OMICS. 2020;24(4):175–9. doi: 10.1089/omi.2020.0019 [DOI] [PubMed] [Google Scholar]
  • 19.Hsieh MH, et al. Liver cancer initiation requires translational activation by an oncofetal regulon involving LIN28 proteins. J Clin Invest. 2024;134(15). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Li M-M, Kong F-E, Li G-M, He Y-T, Zhang X-F, Zhang C-Y, et al. Identification and functional characterization of potential oncofetal targets in human hepatocellular carcinoma. STAR Protoc. 2022;3(4):101921. doi: 10.1016/j.xpro.2022.101921 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Lin H-D, Tseng Y-K, Yuh C-H, Chen S-C. Low concentrations of 4-ABP promote liver carcinogenesis in human liver cells and a zebrafish model. J Hazard Mater. 2022;423(Pt A):126954. doi: 10.1016/j.jhazmat.2021.126954 [DOI] [PubMed] [Google Scholar]
  • 22.Lu J-W, Ho Y-J, Yang Y-J, Liao H-A, Ciou S-C, Lin L-I, et al. Zebrafish as a disease model for studying human hepatocellular carcinoma. World J Gastroenterol. 2015;21(42):12042–58. doi: 10.3748/wjg.v21.i42.12042 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Huang Y, Liu X, Wang H-Y, Chen J-Y, Zhang X, Li Y, et al. Single-cell transcriptome landscape of zebrafish liver reveals hepatocytes and immune cell interactions in understanding nonalcoholic fatty liver disease. Fish Shellfish Immunol. 2024;146:109428. doi: 10.1016/j.fsi.2024.109428 [DOI] [PubMed] [Google Scholar]
  • 24.Tao T, Peng J. Liver development in zebrafish (Danio rerio). J Genet Genomics. 2009;36(6):325–34. doi: 10.1016/S1673-8527(08)60121-6 [DOI] [PubMed] [Google Scholar]
  • 25.Zheng W, Li Z, Nguyen AT, Li C, Emelyanov A, Gong Z. Xmrk, kras and myc transgenic zebrafish liver cancer models share molecular signatures with subsets of human hepatocellular carcinoma. PLoS One. 2014;9(3):e91179. doi: 10.1371/journal.pone.0091179 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Luo J, Lu C, Feng M, Dai L, Wang M, Qiu Y, et al. Cooperation between liver-specific mutations of pten and tp53 genetically induces hepatocarcinogenesis in zebrafish. J Exp Clin Cancer Res. 2021;40(1):262. doi: 10.1186/s13046-021-02061-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Letrado P, et al. Zebrafish: speeding up the cancer drug discovery process. Cancer Res. 2018;78(21):6048–58. [DOI] [PubMed] [Google Scholar]
  • 28.Kim YH, Jeong DC, Pak K, Han M-E, Kim J-Y, Liangwen L, et al. SLC2A2 (GLUT2) as a novel prognostic factor for hepatocellular carcinoma. Oncotarget. 2017;8(40):68381–92. doi: 10.18632/oncotarget.20266 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Wang B, Pu R. Association between glycolysis markers and prognosis of liver cancer: a systematic review and meta-analysis. World J Surg Oncol. 2023;21(1):390. doi: 10.1186/s12957-023-03275-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Zhang X, Li J, Ghoshal K, Fernandez S, Li L. Identification of a subtype of hepatocellular carcinoma with poor prognosis based on expression of genes within the glucose metabolic pathway. Cancers (Basel). 2019;11(12):2023. doi: 10.3390/cancers11122023 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Tonon F, Grassi G. Zebrafish as an experimental model for human disease. Int J Mol Sci. 2023;24(10). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Gallage S, García-Beccaria M, Szydlowska M, Rahbari M, Mohr R, Tacke F, et al. The therapeutic landscape of hepatocellular carcinoma. Med. 2021;2(5):505–52. doi: 10.1016/j.medj.2021.03.002 [DOI] [PubMed] [Google Scholar]
  • 33.Carpentier A, Sheldon J, Vondran FWR, Brown RJ, Pietschmann T. Efficient acute and chronic infection of stem cell-derived hepatocytes by hepatitis C virus. Gut. 2020;69(9):1659–66. doi: 10.1136/gutjnl-2019-319354 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.DeLaForest A, Nagaoka M, Si-Tayeb K, Noto FK, Konopka G, Battle MA, et al. HNF4A is essential for specification of hepatic progenitors from human pluripotent stem cells. Development. 2011;138(19):4143–53. doi: 10.1242/dev.062547 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Irudayam JI, Contreras D, Spurka L, Subramanian A, Allen J, Ren S, et al. Characterization of type I interferon pathway during hepatic differentiation of human pluripotent stem cells and hepatitis C virus infection. Stem Cell Res. 2015;15(2):354–64. doi: 10.1016/j.scr.2015.08.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Kim EK, Yun SJ, Ha JM, Kim YW, Jin IH, Yun J, et al. Selective activation of Akt1 by mammalian target of rapamycin complex 2 regulates cancer cell migration, invasion, and metastasis. Oncogene. 2011;30(26):2954–63. doi: 10.1038/onc.2011.22 [DOI] [PubMed] [Google Scholar]
  • 37.Lachaal M, Rampal AL, Ryu J, Lee W, Hah J, Jung CY. Characterization and partial purification of liver glucose transporter GLUT2. Biochim Biophys Acta. 2000;1466(1–2):379–89. doi: 10.1016/s0005-2736(00)00205-4 [DOI] [PubMed] [Google Scholar]
  • 38.Vogt MC, Hobert O. Starvation-induced changes in somatic insulin/IGF-1R signaling drive metabolic programming across generations. Sci Adv. 2023;9(14):eade1817. doi: 10.1126/sciadv.ade1817 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Liu F, et al. The role of IGF/IGF-1R signaling in the regulation of cancer stem cells. Clin Transl Oncol. 2024;26(12):2924–34. [DOI] [PubMed] [Google Scholar]
  • 40.Kwon EJ, Lee H, Shin U, Kim E-S, Myung K, Kim J, et al. Ionizing radiation inhibits zebrafish embryo hatching through induction of tissue inhibitors of metalloproteinases (TIMPs) expression. FEBS J. 2024;291(24):5470–85. doi: 10.1111/febs.17318 [DOI] [PubMed] [Google Scholar]

Decision Letter 0

Heping Cao

1 Nov 2024

PONE-D-24-37357Solute carrier family 2 member 2 (glucose transporter 2): a common factor of hepatocyte and hepatocellular carcinoma differentiationPLOS ONE

Dear Dr. Kim,

Thank you for submitting your manuscript to PLOS ONE. After careful consideration, we feel that it has merit but does not fully meet PLOS ONE’s publication criteria as it currently stands. Therefore, we invite you to submit a revised version of the manuscript that addresses the points raised during the review process.

==============================

Please address the concerns from the two reviewers.

Please submit your revised manuscript by Dec 16 2024 11:59PM. If you will need more time than this to complete your revisions, please reply to this message or contact the journal office at plosone@plos.org . When you're ready to submit your revision, log on to https://www.editorialmanager.com/pone/ and select the 'Submissions Needing Revision' folder to locate your manuscript file.

Please include the following items when submitting your revised manuscript:

  • A rebuttal letter that responds to each point raised by the academic editor and reviewer(s). You should upload this letter as a separate file labeled 'Response to Reviewers'.

  • A marked-up copy of your manuscript that highlights changes made to the original version. You should upload this as a separate file labeled 'Revised Manuscript with Track Changes'.

  • An unmarked version of your revised paper without tracked changes. You should upload this as a separate file labeled 'Manuscript'.

If you would like to make changes to your financial disclosure, please include your updated statement in your cover letter. Guidelines for resubmitting your figure files are available below the reviewer comments at the end of this letter.

If applicable, we recommend that you deposit your laboratory protocols in protocols.io to enhance the reproducibility of your results. Protocols.io assigns your protocol its own identifier (DOI) so that it can be cited independently in the future. For instructions see: https://journals.plos.org/plosone/s/submission-guidelines#loc-laboratory-protocols . Additionally, PLOS ONE offers an option for publishing peer-reviewed Lab Protocol articles, which describe protocols hosted on protocols.io. Read more information on sharing protocols at https://plos.org/protocols?utm_medium=editorial-email&utm_source=authorletters&utm_campaign=protocols .

We look forward to receiving your revised manuscript.

Kind regards,

Heping Cao, PhD

Academic Editor

PLOS ONE

Journal requirements: When submitting your revision, we need you to address these additional requirements. 1. Please ensure that your manuscript meets PLOS ONE's style requirements, including those for file naming. The PLOS ONE style templates can be found at https://journals.plos.org/plosone/s/file?id=wjVg/PLOSOne_formatting_sample_main_body.pdf and https://journals.plos.org/plosone/s/file?id=ba62/PLOSOne_formatting_sample_title_authors_affiliations.pdf 2. Please match your authorship list in your manuscript file and in the system. 3. To comply with PLOS ONE submissions requirements, in your Methods section, please provide additional information regarding the experiments involving animals and ensure you have included details on (1) methods of sacrifice, (2) methods of anesthesia and/or analgesia, and (3) efforts to alleviate suffering. 4. PLOS ONE now requires that authors provide the original uncropped and unadjusted images underlying all blot or gel results reported in a submission’s figures or Supporting Information files. This policy and the journal’s other requirements for blot/gel reporting and figure preparation are described in detail at https://journals.plos.org/plosone/s/figures#loc-blot-and-gel-reporting-requirements and https://journals.plos.org/plosone/s/figures#loc-preparing-figures-from-image-files. When you submit your revised manuscript, please ensure that your figures adhere fully to these guidelines and provide the original underlying images for all blot or gel data reported in your submission. See the following link for instructions on providing the original image data: https://journals.plos.org/plosone/s/figures#loc-original-images-for-blots-and-gels.   In your cover letter, please note whether your blot/gel image data are in Supporting Information or posted at a public data repository, provide the repository URL if relevant, and provide specific details as to which raw blot/gel images, if any, are not available. Email us at plosone@plos.org if you have any questions. 5. We note that the grant information you provided in the ‘Funding Information’ and ‘Financial Disclosure’ sections do not match.  When you resubmit, please ensure that you provide the correct grant numbers for the awards you received for your study in the ‘Funding Information’ section. 6. Please note that your Data Availability Statement is currently missing [the repository name and/or the DOI/accession number of each dataset OR a direct link to access each database]. If your manuscript is accepted for publication, you will be asked to provide these details on a very short timeline. We therefore suggest that you provide this information now, though we will not hold up the peer review process if you are unable. 7. Please include your full ethics statement in the ‘Methods’ section of your manuscript file. In your statement, please include the full name of the IRB or ethics committee who approved or waived your study, as well as whether or not you obtained informed written or verbal consent. If consent was waived for your study, please include this information in your statement as well.  8. Please include captions for your Supporting Information files at the end of your manuscript, and update any in-text citations to match accordingly. Please see our Supporting Information guidelines for more information: http://journals.plos.org/plosone/s/supporting-information. 

9. Please review your reference list to ensure that it is complete and correct. If you have cited papers that have been retracted, please include the rationale for doing so in the manuscript text, or remove these references and replace them with relevant current references. Any changes to the reference list should be mentioned in the rebuttal letter that accompanies your revised manuscript. If you need to cite a retracted article, indicate the article’s retracted status in the References list and also include a citation and full reference for the retraction notice.

[Note: HTML markup is below. Please do not edit.]

Reviewers' comments:

Reviewer's Responses to Questions

Comments to the Author

1. Is the manuscript technically sound, and do the data support the conclusions?

The manuscript must describe a technically sound piece of scientific research with data that supports the conclusions. Experiments must have been conducted rigorously, with appropriate controls, replication, and sample sizes. The conclusions must be drawn appropriately based on the data presented.

Reviewer #1: Yes

Reviewer #2: Yes

**********

2. Has the statistical analysis been performed appropriately and rigorously?

Reviewer #1: Yes

Reviewer #2: Yes

**********

3. Have the authors made all data underlying the findings in their manuscript fully available?

The PLOS Data policy requires authors to make all data underlying the findings described in their manuscript fully available without restriction, with rare exception (please refer to the Data Availability Statement in the manuscript PDF file). The data should be provided as part of the manuscript or its supporting information, or deposited to a public repository. For example, in addition to summary statistics, the data points behind means, medians and variance measures should be available. If there are restrictions on publicly sharing data—e.g. participant privacy or use of data from a third party—those must be specified.

Reviewer #1: Yes

Reviewer #2: Yes

**********

4. Is the manuscript presented in an intelligible fashion and written in standard English?

PLOS ONE does not copyedit accepted manuscripts, so the language in submitted articles must be clear, correct, and unambiguous. Any typographical or grammatical errors should be corrected at revision, so please note any specific errors here.

Reviewer #1: Yes

Reviewer #2: Yes

**********

5. Review Comments to the Author

Please use the space provided to explain your answers to the questions above. You may also include additional comments for the author, including concerns about dual publication, research ethics, or publication ethics. (Please upload your review as an attachment if it exceeds 20,000 characters)

Reviewer #1: This study investigated the effects of GLUT2 on liver differentiation using in vivo, in vitro, and zebrafish models, in conjunction with big data analysis. Analysis of three datasets (GSE132606, GSE25417, GSE67848) from the Gene Expression Omnibus (GEO) database revealed high expression levels of GLUT2 in patients with hepatocellular carcinoma (HCC). However, I still have related questions that I would like to have explained.

1.The introduction of the role of the SLC2A2 gene needs more description and literature support.

2.Source channels for zebrafish purchases need to be supplemented.

3. The clarity of the images needs to be improved.

4.After knocking out the target gene, the PCR results of other indicators did not change significantly.

5.The article has less experimental content, and it is recommended to add relevant protein experiments.

Reviewer #2: This work uses in vitro, in vivo (zebrafish), and in silico models to examine the role of SLC2A2 in hepatic differentiation and hepatocellular cancer (HCC). According to the findings, SLC2A2 expression and genes linked to stemness are negatively correlated, indicating that it plays a critical role in liver cell differentiation. Targeting SLC2A2 in HCC has therapeutic potential, which is further investigated in this study. Several comments will improve the quality of this study.

Major comments.

1. To better establish whether Slc2a2 influences hepatocyte differentiation, it would be beneficial to analyze additional liver-specific markers. This could provide stronger evidence for the role of Slc2a2 in the differentiation process.

2. The use of zebrafish embryos to explore liver differentiation is an excellent idea. As you have examined the relationship between EGFR, IGF1R, and slc2a2 in in vitro models, it would be interesting to investigate whether similar results are observed in the developing embryos.

3. In Figure 1, it would be helpful to check whether well-established liver differentiation markers (such as albumin, CYP3A4, G6PC) increase as the embryos develop. This would help confirm that the datasets from GEO accurately represent time points in embryonic development.

4. While the study has identified that SLC2A2 plays a role in liver differentiation and cancer progression, it does not fully elucidate the underlying mechanisms behind these effects. This lack of mechanistic clarity represents a limitation of the study. I suggest adding this point to the Discussion section of the manuscript to acknowledge this limitation.

Minor comment.

1. Figures 4C and 4D, please provide a more detailed explanation of what the y-axis represents in Figures 4C and 4D, so that the reader can better understand the data.

**********

6. PLOS authors have the option to publish the peer review history of their article (what does this mean? ). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy .

Reviewer #1: No

Reviewer #2: No

**********

[NOTE: If reviewer comments were submitted as an attachment file, they will be attached to this email and accessible via the submission site. Please log into your account, locate the manuscript record, and check for the action link "View Attachments". If this link does not appear, there are no attachment files.]

While revising your submission, please upload your figure files to the Preflight Analysis and Conversion Engine (PACE) digital diagnostic tool, https://pacev2.apexcovantage.com/ . PACE helps ensure that figures meet PLOS requirements. To use PACE, you must first register as a user. Registration is free. Then, login and navigate to the UPLOAD tab, where you will find detailed instructions on how to use the tool. If you encounter any issues or have any questions when using PACE, please email PLOS at figures@plos.org . Please note that Supporting Information files do not need this step.

PLoS One. 2025 Apr 25;20(4):e0321020. doi: 10.1371/journal.pone.0321020.r003

Author response to Decision Letter 1


21 Jan 2025

Point-to-Point Answers to Reviewers Comments

Reviewer #1

1.The introduction of the role of the SLC2A2 gene needs more description and literature support.

� Thank you for your kind comments. As you suggested, we have elaborated on the function of slc2a2 in the Introduction section to help readers better understand the manuscript. Below is the additional contents (Please see the attached response letter file).

2.Source channels for zebrafish purchases need to be supplemented.

� As per your suggestion, we have made the necessary adjustments. In the Materials and Methods section, we have added that the zebrafish and fabp10a:mCherry transgenic zebrafish embryos used in this study were obtained from the Korea Zebrafish Resource Center (KZRC). Thank you for your valuable input.

3. The clarity of the images needs to be improved.

� We agree with your opinion. Accordingly, we have replaced the image with a higher-resolution version to enhance its clarity and quality.

4.After knocking out the target gene, the PCR results of other indicators did not change significantly.

� We agree with your opinion. To prevent confusion for the readers, we have revised the figure to display only the differentially expressed genes, removing the remaining genes from the figure.

5.The article has less experimental content, and it is recommended to add relevant protein experiments.

� As you suggested, additional experiments seem necessary. Therefore, we utilized the fabp10a:mCherry transgenic zebrafish embryos, which specifically mark the liver, to investigate the effects of slc2a2 knockdown (K/D). Our results showed that the liver size decreased when slc2a2 was knocked down, as confirmed at the protein level (Figure 4G). Additionally, to determine whether IGF1R protein levels also increase upon slc2a2 knockdown, we performed immunohistochemistry (IHC). The IHC results demonstrated that IGF1R expression increased across the whole body when slc2a2 was knocked down (Figure 5C).

Reviewer 2

Major comments.

1. To better establish whether Slc2a2 influences hepatocyte differentiation, it would be beneficial to analyze additional liver-specific markers. This could provide stronger evidence for the role of Slc2a2 in the differentiation process.

� Thank you for your kind suggestion. Based on your comments, we measured the mRNA level of g6pc1a.1, another mature liver marker, in zebrafish embryos. Additionally, we analyzed the mRNA levels of albumin, G6PC, and CYP3A4 using public data. Below are the experimental results: (Please see the attached response letter file)

Figure 1A

Figure 4E

2. The use of zebrafish embryos to explore liver differentiation is an excellent idea. As you have examined the relationship between EGFR, IGF1R, and slc2a2 in in vitro models, it would be interesting to investigate whether similar results are observed in the developing embryos.

� We agree with your opinion and have conducted experiments to investigate this relationship in zebrafish embryos. After performing a knockdown of slc2a2, we observed an increase in the mRNA level of IGF1R, as confirmed by qPCR. Additionally, IHC analysis revealed an increase in the protein level of IGF1R (Figure 5 B and C). These results suggest that slc2a2 may play a regulatory role in modulating IGF1R expression during liver differentiation in zebrafish embryos. Notably, we also examined EGFR expression but did not observe significant changes in either mRNA or protein levels following slc2a2 knockdown. Therefore, our analysis focused on IGF1R, as it demonstrated a more consistent and significant response. These findings further highlight the importance of IGF1R as a key regulatory pathway influenced by slc2a2 in liver differentiation.

3. In Figure 1, it would be helpful to check whether well-established liver differentiation markers (such as albumin, CYP3A4, G6PC) increase as the embryos develop. This would help confirm that the datasets from GEO accurately represent time points in embryonic development.

� Thank you for your kind comments. As per your suggestion, we analyzed the expression levels of albumin, G6PC, and CYP3A4 in datasets GSE132606, GSE25417, and GSE67848. The results demonstrated that the expression of all three genes increased as liver differentiation progressed (Figure 1A).

4. While the study has identified that SLC2A2 plays a role in liver differentiation and cancer progression, it does not fully elucidate the underlying mechanisms behind these effects. This lack of mechanistic clarity represents a limitation of the study. I suggest adding this point to the Discussion section of the manuscript to acknowledge this limitation.

� Thank you for your valuable suggestion. As you recommended, we have included this point in the Discussion section of the manuscript to acknowledge the limitation regarding the lack of mechanistic clarity behind the effects of SLC2A2 on liver differentiation and cancer progression. This addition highlights the need for further studies to fully elucidate the underlying mechanisms.

Attachment

Submitted filename: Point-to-Point Answers to Reviewers Comments.docx

pone.0321020.s003.docx (1.3MB, docx)

Decision Letter 1

Heping Cao

28 Feb 2025

Solute carrier family 2 member 2 (glucose transporter 2): a common factor of hepatocyte and hepatocellular carcinoma differentiation

PONE-D-24-37357R1

Dear Dr. Kim,

We’re pleased to inform you that your manuscript has been judged scientifically suitable for publication and will be formally accepted for publication once it meets all outstanding technical requirements.

Within one week, you’ll receive an e-mail detailing the required amendments. When these have been addressed, you’ll receive a formal acceptance letter and your manuscript will be scheduled for publication.

An invoice will be generated when your article is formally accepted. Please note, if your institution has a publishing partnership with PLOS and your article meets the relevant criteria, all or part of your publication costs will be covered. Please make sure your user information is up-to-date by logging into Editorial Manager at Editorial Manager®  and clicking the ‘Update My Information' link at the top of the page. If you have any questions relating to publication charges, please contact our Author Billing department directly at authorbilling@plos.org.

If your institution or institutions have a press office, please notify them about your upcoming paper to help maximize its impact. If they’ll be preparing press materials, please inform our press team as soon as possible -- no later than 48 hours after receiving the formal acceptance. Your manuscript will remain under strict press embargo until 2 pm Eastern Time on the date of publication. For more information, please contact onepress@plos.org.

Kind regards,

Heping Cao, PhD

Academic Editor

PLOS ONE

Additional Editor Comments (optional):

Reviewers' comments:

Reviewer's Responses to Questions

Comments to the Author

1. If the authors have adequately addressed your comments raised in a previous round of review and you feel that this manuscript is now acceptable for publication, you may indicate that here to bypass the “Comments to the Author” section, enter your conflict of interest statement in the “Confidential to Editor” section, and submit your "Accept" recommendation.

Reviewer #2: All comments have been addressed

**********

2. Is the manuscript technically sound, and do the data support the conclusions?

The manuscript must describe a technically sound piece of scientific research with data that supports the conclusions. Experiments must have been conducted rigorously, with appropriate controls, replication, and sample sizes. The conclusions must be drawn appropriately based on the data presented.

Reviewer #2: Yes

**********

3. Has the statistical analysis been performed appropriately and rigorously?

Reviewer #2: Yes

**********

4. Have the authors made all data underlying the findings in their manuscript fully available?

The PLOS Data policy requires authors to make all data underlying the findings described in their manuscript fully available without restriction, with rare exception (please refer to the Data Availability Statement in the manuscript PDF file). The data should be provided as part of the manuscript or its supporting information, or deposited to a public repository. For example, in addition to summary statistics, the data points behind means, medians and variance measures should be available. If there are restrictions on publicly sharing data—e.g. participant privacy or use of data from a third party—those must be specified.

Reviewer #2: Yes

**********

5. Is the manuscript presented in an intelligible fashion and written in standard English?

PLOS ONE does not copyedit accepted manuscripts, so the language in submitted articles must be clear, correct, and unambiguous. Any typographical or grammatical errors should be corrected at revision, so please note any specific errors here.

Reviewer #2: Yes

**********

6. Review Comments to the Author

Please use the space provided to explain your answers to the questions above. You may also include additional comments for the author, including concerns about dual publication, research ethics, or publication ethics. (Please upload your review as an attachment if it exceeds 20,000 characters)

Reviewer #2: The authors have adequately addressed my comments raised in a previous manuscript. So there is no more comment.

**********

7. PLOS authors have the option to publish the peer review history of their article (what does this mean? ). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy .

Reviewer #2: No

**********

Acceptance letter

Heping Cao

PONE-D-24-37357R1

PLOS ONE

Dear Dr. Kim,

I'm pleased to inform you that your manuscript has been deemed suitable for publication in PLOS ONE. Congratulations! Your manuscript is now being handed over to our production team.

At this stage, our production department will prepare your paper for publication. This includes ensuring the following:

* All references, tables, and figures are properly cited

* All relevant supporting information is included in the manuscript submission,

* There are no issues that prevent the paper from being properly typeset

If revisions are needed, the production department will contact you directly to resolve them. If no revisions are needed, you will receive an email when the publication date has been set. At this time, we do not offer pre-publication proofs to authors during production of the accepted work. Please keep in mind that we are working through a large volume of accepted articles, so please give us a few weeks to review your paper and let you know the next and final steps.

Lastly, if your institution or institutions have a press office, please let them know about your upcoming paper now to help maximize its impact. If they'll be preparing press materials, please inform our press team within the next 48 hours. Your manuscript will remain under strict press embargo until 2 pm Eastern Time on the date of publication. For more information, please contact onepress@plos.org.

If we can help with anything else, please email us at customercare@plos.org.

Thank you for submitting your work to PLOS ONE and supporting open access.

Kind regards,

PLOS ONE Editorial Office Staff

on behalf of

Dr. Heping Cao

Academic Editor

PLOS ONE

Associated Data

    This section collects any data citations, data availability statements, or supplementary materials included in this article.

    Supplementary Materials

    S1 Fig. Protein levels of GLUT2 were analyzed by western blotting.

    GLUT2 protein expression was quantified compared to the control cells., ** p < 0.01, and *** p < 0.001.

    (PPTX)

    pone.0321020.s001.pptx (1.2MB, pptx)
    Attachment

    Submitted filename: Point-to-Point Answers to Reviewers Comments.docx

    pone.0321020.s003.docx (1.3MB, docx)

    Data Availability Statement

    All GSE data files are available from the GEO database (GSE132606, GSE25417, GSE67848).


    Articles from PLOS One are provided here courtesy of PLOS

    RESOURCES