Skip to main content

GATA binding protein 4 promotes the expression and transcription of hepatitis B virus by facilitating hepatocyte nuclear factor 4 alpha in vitro

Abstract

Background

GATA binding protein 4 (GATA4) has been reported as a potential target of gene therapy for hepatocellular carcinoma (HCC). It is well known that the main cause of HCC is the chronic infection of hepatitis B virus (HBV). However, whether the effect of GATA4 on HBV has not yet been reported.

Methods

In this study, the regulation of GATA4 on HBV was analyzed in vitro. In turn, the effect of HBV on GATA4 was also observed in vitro, in vivo, and clinical HCC patients. Subsequently, we analyzed whether the effect of GATA4 on HBV was related to hepatocyte nuclear factor 4 alpha (HNF4α) in vitro.

Results

The results showed that GATA4 significantly promoted the secretion of HBV surface antigen (HBsAg) and HBV e antigen in the cell culture medium, improved the replication of HBV genomic DNA, and increased the level of HBV 3.5 kb pre-genomic RNA and HBV total RNA (P < 0.05). Moreover, it was showed that HBV had no significant effect on GATA4 in vitro and in vivo (P > 0.05). At the same time, GATA4 expression was decreased in 78.9% (15/19) of HCC patients regardless of the HBV and HBsAg status. Among them, there were 76.9% (10/13) in HBV-associated patients with HCC (HBV-HCC), and 83.3% (5/6) in non-HBV-HCC patients. In addition, the expression of HNF4α was also up-regulated or down-regulated accordingly when stimulating or interfering with the expression of GATA4. Furthermore, stimulating the expression of HNF4α could only alleviate the HBsAg level and HBV transcription levels, but had no significant effect on GATA4.

Conclusions

In summary, this study found that GATA4 has a positive effect on HBV, and the potential pathway may be related to another transcription factor HNF4α that regulates HBV.

Introduction

It is reported that the global incidence and mortality of hepatocellular carcinoma (HCC) in 2018 are 4.7% and 8.2% respectively, which is the sixth most common cancer and the fourth leading cause of death for cancer [1]. Treatment methods for HCC include surgical resection, radiotherapy, and chemotherapy, each of with its shortcomings [2]. Gene therapy is considered to be a potential treatment for effectively suppressing HCC with the in-depth research on tumor suppressor genes [3]. GATA binding protein 4 (GATA4), one of the members of the GATA transcription family, is a key transcription factor that regulates cell differentiation and tissue development [4, 5]. Studies have reported that GATA4 can inhibit the development of a variety of cancers [6,7,8,9]. Recently, GATA4 has also been found to be an important tumor suppressor gene for HCC [10, 11]. Previously, we found that GATA4 induces mesenchymal-to-epithelial transition and cellular senescence through the NF-κB pathway in hepatocellular carcinoma. GATA4 plays a crucial tumor-suppressive role in HCC [12].

HCC is a primary liver cancer, and its main pathogenic factors include alcoholism, drug stimulation, obesity, virus (hepatitis B virus and hepatitis C virus), and so on [13]. Chronic hepatitis B (CHB) is the most common pathogenic factor leading to HCC [14,15,16]. HBV chronic infection accounts for at least 50% of HCC patients in the worldwide [14], and even more than to 90% in some regions [17]. Transcription factors play an important role in chronic HBV infection [18]. Interestingly, studies have reported that GATA4 can regulate retrovirus infection [19,20,21]. However, the relationship between GATA4 and HBV is unclear.

Hepatocyte nuclear factor 4 alpha (HNF4α) is a necessary factor that regulates HBV transcriptional activity in hepatocytes. It can bind to the basic core promoter region of HBV cccDNA to promote synthesis of the 3.5 kb pgRNA [22, 23]. The continuous amplification of HBV in host cells relies on the reverse transcription process of HBV using 3.5 kb pgRNA as a template [24]. More importantly, both GATA4 and HNF4α are involved in the regulation of tissue development pathways, and GATA4 is considered to be the potential upstream of HNF4α, regulating different stages of liver development [25]. Whether there is an association between GATA4 and HNF4α in the regulation of HBV remains to be further observed.

In this study, we discovered for the first time that GATA4 may play a positive regulatory role on HBV in vitro. Meanwhile, HBV status was found to have no significant effect on GATA4. Further analysis showed that HNF4α may be located downstream of GATA4 and regulate the level of HBV transcription.

Materials and methods

Retrospective analysis of GATA4 expression and HBV carrying status in clinical HCC patients

19 HCC clinical patient information involved in the previous study was obtained from the Molecular Oncology and Epigenetics Laboratory of the First Affiliated Hospital of Chongqing Medical University (Chongqing, China) [12]. This study retrospectively analyzed the relationship between GATA4 expression and HBV carrier status in these patients.

Cell lines and HBV cccDNA preparation

The normal liver cell line used in this study was L02 cells, the hepatoma or hepatoblastoma cell lines were SMMC7721, HepG2 and HepG2.2.15 cells. These cell lines were purchased from the ATCC cell bank (American Type Culture Collection, Manassas, VA, USA), and all cell lines have been tested for mycoplasma according to the manufacturer’s protocol (Solarbio, Beijing, China). The synthesis of HBV cccDNA was performed as described in a previous study [26]. Briefly, the HBV genome fragment was amplified and synthesized using the phusion high-fidelity DNA polymerase (F530L, Thermo scientific, Lithuania) and the HBV-pEASY plasmid as a template, then subjected to restriction digestion, circularization, and purification to obtain HBV cccDNA.

Cell culture and transfection

L02 cells and SMMC7721 cells were maintained in RPMI 1640 medium (Thermo Fisher Scientific, Suzhou, China) supplemented with 10% fetal bovine serum (Biological Industries, Beit HaEmek, Israel). HepG2 cells and HepG2.2.15 cells were maintained in Dulbecco’s modified Eagle’s medium (Thermo Fisher Scientific) supplemented with 10% fetal bovine serum. All cell lines were treated with 1% penicillin and streptomycin and grown in 5% CO2 at 37 °C. L02 cells and SMMC7721 cells were plated and cultured overnight, then co-transfected with stable expression plasmid of GATA4 (represented as adGATA4) and HBV cccDNA for 48 h (h). HepG2 cells were plated overnight and treated with a concentration of 10 nM GATA4 small interfering RNA (siGATA4) for 24 h, and then transfected with HBV cccDNA for 48 h. HepG2.2.15 cells were only treated with siGATA4 for 48 h. Lipofectamine™ 8000 reagent (Beyotime, Shanghai, China) was used in all transfection processes according to the manufacturer’s protocol. The adGATA4 and siGATA4 are the same as in our previous study [12]. In addition, HepG2.2.15 cells were treated with a specific stimulator of HNF4α, benfluorex hydrochloride (JP-992 hydrochloride; MedChem Express, Monmouth Junction, NJ, USA) for 48 h after siGATA4 for 24 h, the concentration of benfluorex hydrochloride was the same as reported previously [27]. Each experiment was repeated independently at least three times.

Reverse transcription polymerase chain reaction (RT-PCR) and reverse transcription quantitative PCR (RT-qPCR)

Total RNA was extracted using TRIzol reagent (Invitrogen, Carlsbad, CA, USA). Total RNA (1 µg) was reverse transcribed at 42 °C for 1 h into cDNA using GoScript™ Reverse Transcriptase (A5002, Promega, Beijing, China). HBV Genomic DNA was extracted from cells and liver tissues with a TIANamp Genomic DNA kit (TIANGEN, Beijing, Chain). All operations were performed using the manufacturers’ protocols. The mRNA levels of GATA4, HNF4α, HBV 3.5 kb pgRNA, and HBV total RNA were detected by RT-PCR or RT-qPCR, and β-actin was used as the internal control. Copies of HBV DNA from genomic DNA were determined by RT-qPCR with a serial dilution of known copies of HBV standards. The RT-qPCR was conducted using the Bio-Rad CFX connect Real-Time system with iTaq Universal SYBR® Green Supermix (Bio-Rad, Hercules, CA, USA), referring to the manufacturer’s instructions. The 2−∆∆Ct method was used to calculate the Cq data of the samples obtained by RT-qPCR. At least three independent repeat experiments were performed. All primers used are listed in Table 1.

Table.1 Summary of primer sequences used in this study

Enzyme-linked immunosorbent assays (ELISA)

The level of HBsAg and HBeAg in cell culture medium were detected by using ELISA kits according to the manufacturer’s instructions (Kehua Bioengineering, Shanghai, China). The results were judged to be effective or not in advance referencing the instructions. Subsequently, the HBsAg and HBeAg levels in each sample were calculated according to the standard curve established with HBsAg and HBeAg standard (Kangchesitan, Beijing, China).

Western blotting analysis

Total cellular proteins were extracted using radioimmunoprecipitation assay lysis solution (strong; Beyotime), a protease/phosphatase inhibitor mix (Universal Type, 50×; Beyotime), and ultrasonic disruption. After centrifugation at 13,400 ×g for 15 min at 4 °C, the concentration of protein in the supernatant was detected using a bicinchoninic acid protein assay kit (Solarbio, Beijing, China). Then, 20% volume of 6× protein loading buffer (Beyotime) was added to the supernatant, all operations were followed the manufacturer's agreement. The protein was then placed at 100 °C for 5–8 min for denaturation. Subsequently, the protein samples were electrophoretically separated on a 7.5% sodium dodecyl sulfate polyacrylamide gel, then transferred onto a 0.45 μm polyvinylidene fluoride membrane (Merck KGaA, Darmstadt, Germany) in an ice bath. The membrane was blocked with QuickBlock™ Western Blocking Solution (Beyotime) for 15 min, washed three times with tris-buffered saline containing 0.1% Tween 20 for 15 min each time, and the incubated with the primary antibody at 4 °C overnight. The primary antibodies used were as follows: rabbit anti-HNF4α antibody (bs-3828R, BIOSS, Beijing, China; 1:1000), mouse anti-human GATA4 monoclonal antibody (sc-25310, Santa Cruz Biotechnology, Shanghai, China; 1:1000), and mouse anti-tubulin antibody (sc-5274, Santa Cruz Biotechnology, Shanghai, China; 1:1000) as the internal control. After incubation with the primary antibodies, the membrane was washed with tris-buffered saline containing 0.1% Tween 20 and incubated with the anti-rabbit or anti-mouse secondary antibody for 1 h. Proteins were visualized using an ultra-high sensitivity enhanced chemiluminescence kit (HY-K1005, MedChem Express). All assays were performed three times independently.

Immunofluorescence analysis

Cells were seeded into 24-well plates with microcoverslips overnight and then transfected with a GATA4 plasmid and cultured for 48 h. Cells were fixed with 4% paraformaldehyde for 30 min, treated with rupture working solution (PN00014) for 10 min, and blocked with serum for 30 min. And then the slides were incubated with primary antibodies at 4 °C overnight. The primary antibodies used were as follows: GATA4 mouse monoclonal antibody, Clone OTI9F9 (TA500091S, OriGene, Beijing, China; 1:100), HNF4α (C11F12) Rabbit monoclonal antibody (3113S, Cell Signaling Technology, Boston, USA, 1:1500). Subsequently, cells were incubated with Alexa Fluor CY3-conjugated or 488-conjugated goat antimouse or goat anti-rabbit secondary antibodies (Jackson ImmunoResearch, West Grove, PA, USA) for 1 h at 37 C in the dark. Finally, the nuclei were counterstained with 40-6-diamidino-2-phenylindole (DAPI, C0060, Solarbio, Beijing, China) in the dark for 10 min. Micrographs were captured by using fluorescence microscopy. Assays were performed in three separate times.

Statistical analysis

Data were analyzed using SPSS 22.0 software (IBM, Chicago, IL, USA) and ImageJ v1.8.0 (NIH, Bethesda, Maryland, USA). Continuous variables are reported as means ± standard deviations calculated by Prism Software version 5.0 (GraphPad Software, La Jolla, CA, USA). Data with a normal distribution were compared with a two-tailed Student’s t-test. All experiments were repeated independently at least three times. Significance was set at P < 0.05 (non-significance at P ≥ 0.05).

Results

Expression of GATA4 in cells

We first verified the expression of GATA4 in different normal liver cell lines or HCC cell lines via RT-PCR and western blotting (WB) to determine the appropriate cell models. The mRNA levels (Fig. 1a) and protein levels (Fig. 1b) results of these cells showed that GATA4 was expressed at low levels in the L02 cells and SMMC7721 cells, but at highly levels in the HepG2 cells and HepG2.2.15 cells. The expression level of GATA4 in different cells was the basis for the selection of cell models. Therefore, in this study, plasmid adGATA4 was treated in L02 cells and SMMC7721 cells, and siGATA4 was interfered in HepG2 cells and HepG2.2.15 cells.

Fig. 1
figure 1

The expression of GATA4 in hepatocyte lines. a GATA4 mRNA level in cells detected via reverse-transcription PCR (RT-PCR). b Protein of GATA4 (51 kDa) analyzed by western blotting (WB), and tubulin (55 kDa) was used as a loading control

GATA4 overexpression enhances the levels of HBV DNA, HBsAg and HBeAg, 3.5 kb pgRNA and total RNA

To verify whether GATA4 has a regulatory effect on HBV, adGATA4 with different mass concentrations and equal amounts of HBV cccDNA were co-transfected into L02 cells and SMMC7721 cells, respectively. The results showed that the expression of GATA4 increased with the increasing adGATA4 concentrations (all P < 0.01, Fig. 2a). The levels of HBV DNA copies in the cells increased significantly in the treatment group with high concentration of adGATA4 (Fig. 2b). The levels of HBsAg and HBeAg secreted into the culture medium were consistent with GATA4 expression, the increase of HBsAg was particularly significant (all P < 0.05, Fig. 2c).

Fig. 2
figure 2

Overexpression of GATA4 may promote HBV in L02 cells and SMMC7721 cells, respectively. L02 cells and SMMC7721 cells were transfected HBV cccDNA (500 ng per 1 mL 1640 medium, as 500 ng/mL) and adGATA4 for 48 h. (Description for the final concentration of adGATA4: − , control; + , 100 ng/mL; ++ , 200 ng/mL; +++ , 400 ng/mL.) a Relative expression of GATA4 mRNA in cells detected via RT-qPCR, β-actin was used as the internal control. b Analysis of HBV DNA copies in cells determined by qPCR. c HBsAg and HBeAg levels in the culture medium detected through enzyme-linked immunosorbent assays. d Relative levels of HBV 3.5 kb pgRNA and total RNA in cells analyzed via RT-qPCR, β-actin was used as the internal control. e Protein of GATA4 (51 kDa) analyzed via WB. All statistics are marked as NS P > 0.05, *P < 0.05, **P < 0.01, ***P < 0.001

Then, the levels of 3.5 kb pgRNA and total RNA of HBV were further observed after transfection with adGATA4 with a final concentration of 400 ng/mL. The results showed that the levels of 3.5 kb pgRNA and total RNA of HBV were increased compared with the normal group (Fig. 2d), although the increase in the level of HBV 3.5 kb pgRNA in L02 cells was not significant (P > 0.05). GATA4 protein was increased after transfection with adGATA4 (Fig. 2e). General results revealed that GATA4 may be involved in promoting the expression and replication of HBV.

siGATA4 negatively regulates HBV

To further verify the effect of GATA4 on HBV, we silenced GATA4 expression in HepG2 cells and HepG2.2.15 cells using siGATA4. The expression of GATA4 was effectively suppressed in the siGATA4 group (all P < 0.001, Fig. 3a). The levels of HBV DNA and HBsAg were reduced in cells (Fig. 3b, c). Moreover, the levels of 3.5 kb pgRNA and total RNA of HBV were significantly decreased after siGATA4 (all P < 0.01, Fig. 3d). GATA4 protein was also decreased after transfection with siGATA4 (Fig. 3e). The above results converse to prove the promoting effect of GATA4 on HBV.

Fig. 3
figure 3

siGATA4 negatively regulates HBV. HepG2 cells were transfected with siGATA4 (10 nM) for 24 h, and then transfected with HBV cccDNA (500 ng/mL) for 48 h. HepG2.2.15 cells carry the complete genome of HBV, so these were only transfected with siGATA4 for 48 h. a Relative mRNA of GATA4 in cells detected via RT-qPCR, β-actin was used as the internal control. b Analysis of HBV DNA copies in cells determined by qPCR. c HBsAg levels in the culture medium detected through enzyme-linked immunosorbent assays. d Relative levels of HBV 3.5 kb pgRNA and total RNA in cells analyzed via RT-qPCR, β-actin was used as the internal control. e Protein of GATA4 (51 kDa) analyzed via WB. All statistics are marked as NS P > 0.05, *P < 0.05, **P < 0.01, ***P < 0.001

GATA4 may be an upstream transcription factor regulating HBV

In order to observe whether HBV affected the expression of GATA4, we used HBV cccDNA cell model to observe the mRNA level of GATA4, and the results showed there was no clear difference in GATA4 expression in HepG2 cells before and after transfection with HBV (P = 0.46, Fig. 4a). Subsequently, the copies of HBV DNA and the mRNA level of GATA4 were analyzed in liver tissue samples of the HBV mouse models at 4 and 24 weeks [26]. The results showed that the average copy numbers of HBV DNA are 4.2 × 104 copies and 5.7 × 104 copies in the liver tissue of the HBV CBA/CaJ mice (Fig. 4b). The expression of GATA4 did not significantly change in HBV models group compared with the control mice (P = 0.91 or P = 0.26, Fig. 4c). The results demonstrated that HBV had no significant effect on GATA4 in HBV cccDNA cell models and mouse models.

Fig. 4
figure 4

HBV had no significant effect on GATA4 in HBV cccDNA cell model and mouse model. HepG2 cells were transfected with HBV cccDNA (500 ng/mL) for 48 h. a Relative mRNA of GATA4 in cells detected via RT-qPCR, β-actin was used as the internal control. P = 0.46 vs. control group. b Analysis of HBV DNA copies in liver of HBV CBA/CaJ mouse models determined by qPCR, the average copy numbers of HBV DNA are 4.2 × 104 copies and 5.7 × 104 copies at 4 weeks and 24 weeks, respectively. c Relative mRNA of GATA4 in liver of HBV CBA/CaJ mouse models (n = 3) detected via RT-qPCR, β-actin was used as the internal control. P = 0.91 or P = 0.26 vs. control group

GATA4 reduced expression in HCC patients regardless of the HBV and HBsAg status of patients

The levels of GATA4 in clinical patients (n = 19) were based on the results of the previous study [12]. The results showed that 68.4% of the patients were HBV and HBsAg positive, and 31.6% were HBV and HBsAg negative in the 19 HCC patients. Compared with adjacent tissues, the high or low expression of GATA4 in cancer tissues accounted for 23.1% and 76.9%, 16.7% and 83.3% in HBV-HCC patients and in non-HBV-HCC patients, respectively. The results showed that regardless of the patient's HBV and HBsAg status, the expression level of GATA4 decreased in liver cancer tissues of most patients (Table 2).

Table.2 Retrospective analysis of GATA4 expression and HBV carrying status in clinical HCC patients

GATA4 may be associated with the host factor HNF4α that regulates HBV transcription

To observe whether HNF4α is related to the regulation of HBV by GATA4, HNF4α was detected in adGATA4 or siGATA4 cell models using RT-qPCR or western blotting (WB) assay. The results suggested that the mRNA and protein levels of HNF4α were concomitant with the expression of GATA4 (Fig. 5a, b). The results of immunofluorescence analysis of GATA4 and HNF4α in cells showed that, compared with the control group, the expression of HNF4α was increased or decreased in the cells transfected with adGATA4 or siGATA4 (Fig. 5c). Collectively, these results indicate that GATA4 may be associated with the host factor HNF4α.

Fig. 5
figure 5

The relationship between GATA4 and HNF4α in cells. SMMC7721 cells and HepG2.2.15 cells were transfected with adGATA4 or siGATA4 for 48 h. a, b Relative mRNA and protein of HNF4α in cells measured by RT-qPCR and WB. Protein of GATA4 (51 kDa) and HNF4α (52 kDa) were analyzed by WB, and tubulin (55 kDa) was used as a loading control. All statistics are marked as NS P > 0.05, *P < 0.05, **P < 0.01, ***P < 0.001. c Immunofluorescence was used to evaluate the relationship between GATA4 and HNF4α

Stimulating HNF4α alleviates the inhibitory effect of siGATA4 on HBV in HepG2.2.15 cells

To further confirm the role of HNF4α in HBV regulation by GATA4, GATA4 and HNF4α were observed in HepG2.2.15 cells. The results displayed that HNF4α levels could be specifically upregulated by benfluorex hydrochloride but had no significant effect on GATA4 in HepG2.2.15 cells (Fig. 6a, b). Meanwhile, HBsAg and HBV RNAs were significantly reversed by benfluorex hydrochloride in siGATA4 HepG2.2.15 cells (Fig. 6c, d). The upregulation of HNF4α attenuated the inhibitory effect of siGATA4 on HBV. Taken together, the overall results revealed that GATA4 may play a role in the upstream of HNF4α in HBV-related pathways (Fig. 7).

Fig. 6
figure 6

Stimulating HNF4α alleviates the inhibitory effect of siGATA4 on HBV in HepG2.2.15 cells. HepG2.2.15 cells were treated with benfluorex hydrochloride (Final concentration was 1 nM), a specific stimulator of HNF4α, for 48 h after siGATA4 for 24 h. a Chemical structure of the HNF4α specific stimulator, benfluorex hydrochloride (JP-992 hydrochloride). b Relative mRNA of GATA4 detected by RT-qPCR. NS, P > 0.05 vs. control of siGATA4. c HBsAg levels in the culture medium measured via ELISA kit assay. d Relative levels of HBV 3.5 kb pgRNA and total RNA detected via RT-qPCR, β-actin was used as the internal control

Fig. 7
figure 7

A possible pathway of GATA4 regulation HBV, GATA4 may promote HBV activity through up-regulating HNF4α

Discussion

HBV is one of the smallest hepatotropic DNA viruses among all identified to date, and HBV infection is one of the most difficult to be eliminated completely [28, 29]. The possibility of HBV being reactivated is still extremely high after receiving anti-tumor treatment, even in HCC patients whose HBV has turned negative [30,31,32]. HBV transcription, which uses HBV covalently closed circular DNA (cccDNA) as a template to synthesize 3.5 kb pgRNA and multiple messenger RNAs, is an important process for HBV persistent infection [33, 34]. This process depends on the activities of various transcription factors and chromatin-modifying enzymes [35, 36]. Therefore, exploring the effect of GATA4 on HBV has significant research value for the treatment of HBV-HCC.

The synthesis of various antigens in HBV utilizes different messenger RNA as the template, among which the HBsAg is the main serum marker of HBV [37]. The levels of HBsAg and HBV DNA are important indicators for judging the active status of HBV replication [38]. 3.5 kb pgRNA is the template for reverse transcription synthesis of HBV genome relaxes circular DNA (rcDNA), so it is a key intermediate to ensure HBV continuous replication [34]. The most important finding of this study was that GATA4 had a positive regulatory effect on HBV. GATA4 overexpression promoted the secretion of HBsAg, increased the copies of HBV DNA, and enhanced the synthesis of HBV 3.5 kb pgRNA and total RNA. In contrast, the levels of HBsAg, HBV DNA and HBV RNAs were downregulated by siGATA4. Although the present study found that GATA4 could promote HBV, it has not yet been verified by southern blotting or northern blotting.

HNF4α is widely regarded as one of the key factors in the activation of HBV cccDNA and the synthesis of pgRNA [23, 39,40,41]. Previous studies found that HNF4α binds to the HBV genome enhancer, IEnhI/Xp [23, 42]. Moreover, HNF4α participates in the formation of cccDNA chromatin microsomes by binding to the core promoter group region of cccDNA, initiating HBV replication and transcription [22, 43]. HNF4α is also a potential target for the treatment of HBV [44,45,46,47,48]. In this study, the results demonstrated that HNF4α was increased simultaneously after the up-regulation of GATA4. In contrast, up-regulation of HNF4α could only increase HBV markers, but had no effect on GATA4. These results suggest that GATA4 may be a potential upstream regulator of HNF4α, and a stimulus of HBV replication and transcription. Remarkably, the direct interaction between GATA4 and HNF4α in this study had not be identified using a luciferase reporter assay and an immunoprecipitation assay. Therefore, the underlying mechanism requires further investigation.

Conclusion

Previous studies have found that GATA4 is a potential anti-HCC therapeutic target, the current study found that the expression of GATA4 was also decreased in cancer tissues of the most HBV-HCC patients. Furthermore, we innovatively found that GATA4 can promote HBV replication and transcription, and the regulation effect of GATA4 on HBV may be related to HNF4α. The overall results imply that the combination of anti-HBV agents should be considered in HBV-HCC patients when GATA4 is used as the target of antitumor gene therapy.

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

Abbreviations

HBV:

Hepatitis B virus

HBsAg:

Hepatitis B surface antigen

HBV DNA:

Hepatitis B virus DNA

cccDNA:

Covalently closed circular DNA

pgRNA:

Pre-genomic RNA

HBV RNA:

Hepatitis B virus RNA

HCC:

Hepatocellular carcinoma

GATA4:

GATA binding protein 4

HNF4α:

Hepatocyte nuclear factor 4 alpha

adGATA4:

Expression plasmid of GATA4

siGATA4:

GATA4 small interfering RNA

References

  1. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68:394–424.

    Article  PubMed  Google Scholar 

  2. De Mattia E, Cecchin E, Guardascione M, Foltran L, Di Raimo T, Angelini F, D’andrea M, Toffoli G. Pharmacogenetics of the systemic treatment in advanced hepatocellular carcinoma. World J Gastroenterol. 2019;25:3870–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Liu YX, Li QZ, Cao YN, Zhang LQ. Identification of key genes and important histone modifications in hepatocellular carcinoma. Comput Struct Biotechnol J. 2020;18:2657–69.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Holtzinger A, Evans T. Gata4 regulates the formation of multiple organs. Development. 2005;132:4005–14.

    Article  CAS  PubMed  Google Scholar 

  5. Watt AJ, Zhao R, Li J, Duncan SA. Development of the mammalian liver and ventral pancreas is dependent on GATA4. BMC Dev Biol. 2007;7:37.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  6. Gao L, Hu Y, Tian Y, Fan Z, Wang K, Li H, Zhou Q, Zeng G, Hu X, Yu L, Zhou S, Tong X, Huang H, Chen H, Liu Q, Liu W, Zhang G, Zeng M, Zhou G, He Q, Ji H, Chen L. Lung cancer deficient in the tumor suppressor GATA4 is sensitive to TGFBR1 inhibition. Nat Commun. 2019;10:1665.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  7. Gong Y, Zhang L, Zhang A, Chen X, Gao P, Zeng Q. GATA4 inhibits cell differentiation and proliferation in pancreatic cancer. PLoS ONE. 2018;13:e0202449.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  8. Ma X, Ji L. Effect of GATA4 gene methylation on proliferation and apoptosis of SGC-7901 gastric cancer cells. Neoplasma. 2020;67:1193–203.

    Article  CAS  PubMed  Google Scholar 

  9. Han X, Tang J, Chen T, Ren G. Restoration of GATA4 expression impedes breast cancer progression by transcriptional repression of ReLA and inhibition of NF-kappaB signaling. J Cell Biochem. 2019;120:917–27.

    Article  CAS  PubMed  Google Scholar 

  10. Lu F, Zhou Q, Liu L, Zeng G, Ci W, Liu W, Zhang G, Zhang Z, Wang P, Zhang A, Gao Y, Yu L, He Q, Chen L. A tumor suppressor enhancing module orchestrated by GATA4 denotes a therapeutic opportunity for GATA4 deficient HCC patients. Theranostics. 2020;10:484–97.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Enane FO, Shuen WH, Gu X, Quteba E, Przychodzen B, Makishima H, Bodo J, Ng J, Chee CL, Ba R, Seng Koh L, Lim J, Cheong R, Teo M, Hu Z, Ng KP, Maciejewski J, Radivoyevitch T, Chung A, Ooi LL, Tan YM, Cheow PC, Chow P, Chan CY, Lim KH, Yerian L, Hsi E, Toh HC, Saunthararajah Y. GATA4 loss of function in liver cancer impedes precursor to hepatocyte transition. J Clin Invest. 2017;127:3527–42.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Xiang Q, Zhou D, He X, Fan J, Tang J, Qiu Z, Zhang Y, Qiu J, Xu Y, Lai G. The zinc finger protein GATA4 induces mesenchymal-to-epithelial transition and cellular senescence through the nuclear factor-kappaB pathway in hepatocellular carcinoma. J Gastroenterol Hepatol. 2019;34:2196–205.

    Article  CAS  PubMed  Google Scholar 

  13. Marengo A, Rosso C, Bugianesi E. Liver cancer: connections with obesity, fatty liver, and cirrhosis. Annu Rev Med. 2016;67:103–17.

    Article  CAS  PubMed  Google Scholar 

  14. Xie Y, Hepatitis B. Virus-associated hepatocellular carcinoma. Adv Exp Med Biol. 2017;1018:11–21.

    Article  CAS  PubMed  Google Scholar 

  15. Sayiner M, Golabi P, Younossi ZM. Disease burden of hepatocellular carcinoma: a global perspective. Dig Dis Sci. 2019;64:910–7.

    Article  CAS  PubMed  Google Scholar 

  16. Jia L, Gao Y, He Y, Hooper JD, Yang P. HBV induced hepatocellular carcinoma and related potential immunotherapy. Pharmacol Res. 2020;159:104992.

    Article  CAS  PubMed  Google Scholar 

  17. Wong MCS, Huang JLW, George J, Huang J, Leung C, Eslam M, Chan HLY, Ng SC. The changing epidemiology of liver diseases in the Asia-Pacific region. Nat Rev Gastroenterol Hepatol. 2019;16:57–73.

    Article  PubMed  Google Scholar 

  18. Oropeza CE, Tarnow G, Sridhar A, Taha TY, Shalaby RE, Mclachlan A. The regulation of HBV transcription and replication. Adv Exp Med Biol. 2020;1179:39–69.

    Article  CAS  PubMed  Google Scholar 

  19. Shulzhenko N, Morgun A, Hsiao W, Battle M, Yao M, Gavrilova O, Orandle M, Mayer L, Macpherson AJ, Mccoy KD, Fraser-Liggett C, Matzinger P. Crosstalk between B lymphocytes, microbiota and the intestinal epithelium governs immunity versus metabolism in the gut. Nat Med. 2011;17:1585–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Yamagata T, Mitani K, Ueno H, Kanda Y, Yazaki Y, Hirai H. Triple synergism of human T-lymphotropic virus type 1-encoded tax, GATA-binding protein, and AP-1 is required for constitutive expression of the interleukin-5 gene in adult T-cell leukemia cells. Mol Cell Biol. 1997;17:4272–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Ito J, Sugimoto R, Nakaoka H, Yamada S, Kimura T, Hayano T, Inoue I. Systematic identification and characterization of regulatory elements derived from human endogenous retroviruses. PLoS Genet. 2017;13:e1006883.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  22. Kim DH, Kang HS, Kim KH. Roles of hepatocyte nuclear factors in hepatitis B virus infection. World J Gastroenterol. 2016;22:7017–29.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Cho EY, Kim HJ, Park C, So HS, Park RK, Kim HC. Impact of nucleotide mutations at the HNF3- and HNF4-binding sites in enhancer 1 on viral replication in patients with chronic hepatitis B virus infection. Gut Liver. 2013;7:569–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Datta S, Chatterjee S, Veer V, Chakravarty R. Molecular biology of the hepatitis B virus for clinicians. J Clin Exp Hepatol. 2012;2:353–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Collins DP, Hapke JH, Aravalli RN, Steer CJ. In vitro differentiation of human TERT-transfected multi-lineage progenitor cells (MLPC) into immortalized hepatocyte-like cells. Hepat Med. 2020;12:79–92.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Liu Y, Zhao ZH, Lv XQ, Tang YW, Cao M, Xiang Q, Wu Y, Zhang HT, Lai GQ. Precise analysis of the effect of basal core promoter/precore mutations on the main phenotype of chronic hepatitis B in mouse models. J Med Virol. 2020. https://doi.org/10.1002/jmv.26025.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Lee SH, Athavankar S, Cohen T, Piran R, Kiselyuk A, Levine F. Identification of alverine and benfluorex as HNF4alpha activators. ACS Chem Biol. 2013;8:1730–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Dandri M, Petersen J. cccDNA maintenance in chronic hepatitis B—targeting the matrix of viral replication. Infect Drug Resist. 2020;13:3873–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Yuen MF, Chen DS, Dusheiko GM, Janssen HLA, Lau DTY, Locarnini SA, Peters MG, Lai CL. Hepatitis B virus infection. Nat Rev Dis Primers. 2018;4:18035.

    Article  PubMed  Google Scholar 

  30. Xie ZB, Zhu SL, Peng YC, Chen J, Wang XB, Ma L, Bai T, Xiang BD, Li LQ, Zhong JH. Postoperative hepatitis B virus reactivation and surgery-induced immunosuppression in patients with hepatitis B-related hepatocellular carcinoma. J Surg Oncol. 2015;112:634–42.

    Article  CAS  PubMed  Google Scholar 

  31. Wang XB, Chen J, Xiang BD, Wu FX, Li LQ. High CONUT score predicts poor survival and postoperative HBV reactivation in HBV-related hepatocellular carcinoma patients with low HBV-DNA levels. Eur J Surg Oncol. 2019;45:782–7.

    Article  PubMed  Google Scholar 

  32. Xu M, Zhou Z, Xu R, Zhang H, Lin N, Zhong Y. Antiviral therapy predicts the outcomes following resection of hepatocellular carcinoma in patients negative for HBV DNA: a propensity score matching analysis. World J Surg Oncol. 2019;17:45.

    Article  PubMed  PubMed Central  Google Scholar 

  33. Dezhbord M, Lee S, Kim W, Seong BL, Ryu WS. Characterization of the molecular events of covalently closed circular DNA synthesis in de novo Hepatitis B virus infection of human hepatoma cells. Antiviral Res. 2019;163:11–8.

    Article  CAS  PubMed  Google Scholar 

  34. Schadler S, Hildt E. HBV life cycle: entry and morphogenesis. Viruses. 2009;1:185–209.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Zhang Y, Mao R, Yan R, Cai D, Zhang Y, Zhu H, Kang Y, Liu H, Wang J, Qin Y, Huang Y, Guo H, Zhang J. Transcription of hepatitis B virus covalently closed circular DNA is regulated by CpG methylation during chronic infection. PLoS ONE. 2014;9:e110442.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  36. Liu F, Campagna M, Qi Y, Zhao X, Guo F, Xu C, Li S, Li W, Block TM, Chang J, Guo JT. Alpha-interferon suppresses hepadnavirus transcription by altering epigenetic modification of cccDNA minichromosomes. PLoS Pathog. 2013;9:e1003613.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Kao JH. Diagnosis of hepatitis B virus infection through serological and virological markers. Expert Rev Gastroenterol Hepatol. 2008;2:553–62.

    Article  CAS  PubMed  Google Scholar 

  38. Gish RG, Given BD, Lai CL, Locarnini SA, Lau JY, Lewis DL, Schluep T. Chronic hepatitis B: virology, natural history, current management and a glimpse at future opportunities. Antiviral Res. 2015;121:47–58.

    Article  CAS  PubMed  Google Scholar 

  39. Zheng Y, Li J, Ou JH. Regulation of hepatitis B virus core promoter by transcription factors HNF1 and HNF4 and the viral X protein. J Virol. 2004;78:6908–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Leng XH, Chen EQ, Du LY, Bai L, Gong DY, Cheng X, Huang FJ, Tang H. Biological characteristics of the A1762T/G1764A mutant strain of hepatitis B virus in vivo. Mol Med Rep. 2015;12:5141–8.

    Article  CAS  PubMed  Google Scholar 

  41. Oropeza CE, Li L, Mclachlan A. Differential inhibition of nuclear hormone receptor-dependent hepatitis B virus replication by the small heterodimer partner. J Virol. 2008;82:3814–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Bock CT, Malek NP, Tillmann HL, Manns MP, Trautwein C. The enhancer I core region contributes to the replication level of hepatitis B virus in vivo and in vitro. J Virol. 2000;74:2193–202.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Chen EQ, Sun H, Feng P, Gong DY, Liu C, Bai L, Yang WB, Lei XZ, Chen LY, Huang FJ, Tang H. Study of the expression levels of Hepatocyte nuclear factor 4 alpha and 3 beta in patients with different outcome of HBV infection. Virol J. 2012;9:23.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. Tseng YP, Wu YC, Leu YL, Yeh SF, Chou CK. Scutellariae radix suppresses hepatitis B virus production in human hepatoma cells. Front Biosci (Elite Ed). 2010;2:1538–47.

    Google Scholar 

  45. Tseng YP, Kuo YH, Hu CP, Jeng KS, Janmanchi D, Lin CH, Chou CK, Yeh SF. The role of helioxanthin in inhibiting human hepatitis B viral replication and gene expression by interfering with the host transcriptional machinery of viral promoters. Antivir Res. 2008;77:206–14.

    Article  CAS  PubMed  Google Scholar 

  46. Bai L, Nong Y, Shi Y, Liu M, Yan L, Shang J, Huang F, Lin Y, Tang H. Luteolin inhibits hepatitis B virus replication through extracellular signal-regulated kinase-mediated down-regulation of hepatocyte nuclear factor 4alpha expression. Mol Pharm. 2016;13:568–77.

    Article  CAS  PubMed  Google Scholar 

  47. Huang H, Zhou W, Zhu H, Zhou P, Shi X. Baicalin benefits the anti-HBV therapy via inhibiting HBV viral RNAs. Toxicol Appl Pharmacol. 2017;323:36–43.

    Article  CAS  PubMed  Google Scholar 

  48. Dai XQ, Cai WT, Wu X, Chen Y, Han FM. Protocatechuic acid inhibits hepatitis B virus replication by activating ERK1/2 pathway and down-regulating HNF4alpha and HNF1alpha in vitro. Life Sci. 2017;180:68–74.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was supported by the National Natural Science Foundation of China [Grant No. 81570541], the Min-Sheng Project of Chongqing Science and Technology [Grant No. cstc2017shmsA130097], and the Chongqing Postgraduate Innovation Project in Scientific Research [Grant No. CYS19219].

Author information

Authors and Affiliations

Authors

Contributions

GQL and XQL designed the research methods and analyzed the data. XQL, XX, YW and RQX were involved in the in vitro cell experiments and drafting the manuscript. YL provided samples and data analysis obtained from in vivo experiments. QX collected the information of 19 HCC clinical patients, and provided experimental materials, such as GATA4 expression plasmid (adGATA4) and small interfering RNA (siGATA4). All authors read and approved the final manuscript and agreed to be accountable for all aspects of the research in ensuring that the accuracy or integrity of any part of the work are appropriately investigated and resolved. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Guoqi Lai.

Ethics declarations

Ethics approval and consent to participate

Not applicable. The analysis of clinical sample information in this study is a retrospective analysis of published research content. The samples and data of the mouse model in this study are derived from the remaining parts of the published research content.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lv, X., Xiang, X., Wu, Y. et al. GATA binding protein 4 promotes the expression and transcription of hepatitis B virus by facilitating hepatocyte nuclear factor 4 alpha in vitro. Virol J 18, 196 (2021). https://doi.org/10.1186/s12985-021-01668-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12985-021-01668-z

Keywords