Hepatitis B virus (HBV) is the most common chronic viral infection globally, affecting ∼360 million people and causing about 1 million deaths annually due to end-stage liver disease or hepatocellular carcinoma. Current antiviral treatments rarely achieve a functional cure for chronic hepatitis B, highlighting the need for improved monitoring and intervention strategies. This study explores the role of the sphingosine kinase 1 (SphK1)–sphingosine-1-phosphate (S1P) axis in HBV-related liver injury. We investigated the association between serum S1P concentration and HBV DNA levels in chronic hepatitis B patients, finding a significant positive correlation. Additionally, SphK1 was elevated in liver tissues of HBV-positive hepatocellular carcinoma patients, particularly in HBsAg-positive regions. HBV infection models in HepG2–sodium taurocholate cotransporting polypeptide cells confirmed that HBV enhances SphK1 expression and S1P production. Inhibition of HBV replication through antiviral agents and the CRISPR-Cas9 system reduced SphK1 and S1P levels. Further, we identified the transcription factor USF1 as a key regulator of SphK1 expression during HBV infection. USF1 binds to the SphK1 promoter, increasing its transcriptional activity, and is upregulated in response to HBV infection. In vivo studies in mice demonstrated that HBV exposure promotes the expression of USF1 and SphK1–S1P. These findings suggest that the SphK1–S1P axis, regulated by HBV-induced USF1, could serve as a potential biomarker and therapeutic target for HBV-related liver injury.

Hepatitis B virus (HBV) is the most common chronic viral infection globally, affecting ∼360 million people, with around one million deaths annually due to end-stage liver disease or hepatocellular carcinoma (HCC) (1). Current antiviral therapies, mainly consisting of IFN and nucleotide analogs, rarely achieve a functional cure for chronic hepatitis B (CHB), defined by seronegative conversion of HBsAg (2). Consequently, increasing research focuses on monitoring and timely intervention to delay HBV-induced liver injury.

The hepatitis B virion is an enveloped nucleocapsid with an outer lipoprotein envelope and an inner nucleocapsid core, which selectively enters hepatocytes by binding to the sodium taurocholate cotransporting polypeptide (NTCP) receptor on the cell surface (3). The HBV genome, a 3.2-kb partially double-stranded circular DNA, contains four overlapping open reading frames (ORFs): 1) ORF-P encoding HBV polymerase (P); 2) pre-S1/pre-S2/S ORF encoding large/middle/small hepatitis B surface Ag (HBsAg); 3) pre-C/C–ORF coding for hepatitis B e Ag (HBeAg) and core protein (HBc); and 4) X-ORF coding for the HBx protein. Each of these proteins plays a distinct role in the HBV life cycle and disease etiology. Immune-mediated host–virus interactions significantly influence the outcome of HBV infection (4, 5). Thus, considerable research aims to alleviate immune injury and hepatocellular carcinogenesis in CHB by targeting the essential immune checkpoints.

Sphingosine-1-phosphate (S1P) is a bioactive lipid metabolite that stimulates DNA synthesis and regulates cell survival, migration, and adhesion by binding to 5 G protein–coupled receptors (S1PR1 to S1PR5) (6, 7). In addition, S1P–S1PR signaling regulates B and T lymphocytes trafficking from lymph nodes to tissues through the circulatory system (8). Sphingosine kinase (SphK) is the key rate‐limiting enzyme in S1P synthesis, existing as two isoforms: SphK1 and SphK2. In mammals, both SphK1 and SphK2 catalyze the conversion of sphingosine (Sph) to S1P, but SphK2 preferentially synthesizes dihydro-S1P (dhS1P) from dihydrosphingosine (dhSph) (9). SphK1 is predominantly found in the cytoplasm, whereas SphK2 is located in the nucleus and in the inner membrane of the Golgi, endoplasmic reticulum, and mitochondria (10). Recent reports have shown that SphK1 is involved in various liver injury processes, including hepatocyte inflammatory necrosis, liver fibrosis, hepatocarcinogenesis, and acute liver failure (11–13). Consequently, many drugs targeting SphK–S1P–S1PR signaling have been developed for various immune-mediated diseases. For instance, a specific SphK1 inhibitor has been shown to ameliorate inflammation in acute liver failure (14), and fingolimod (FTY720), an oral drug targeting downstream S1P–S1PR signaling, has been approved by the Food and Drug Administration for the treatment of multiple sclerosis (15).

SphK1 has been reported to be abnormally expressed in HBV-related liver diseases such as HCC and liver fibrosis (16, 17), but its direct association with HBV has not been confirmed in humans. The current study provides new insights into the interactions between HBV infection and the SphK1–S1P axis. Using human samples, in vivo experiments, and in vitro experiments, we validate the correlation between S1P levels and HBV infection and preliminarily explore the underlying mechanisms. Our findings support the notion that HBV can promote SphK1 expression to release more S1P. Beyond serving as a potential indictor of altered metabolism of HBV-infected hepatocytes, SphK1–S1P–S1PR signal might be a therapeutic target for delaying HBV-related liver injury.

In this trial, 20 patients were diagnosed with hepatitis B according to the American Association for the Study of Liver Diseases 2018 hepatitis B guidance (18), including 11 CHB, 5 hepatitis B–related HCC, 4 and HCC uncertainly caused by hepatitis B. They were treated with nucleos(t)ide analogs (NAs) (entecavir and tenofovir) for more than 3 months according to the doctor’s advice, in the Fourth Affiliated Hospital of Soochow University, China, from 2021 to 2023 (15 male and 5 females; age range, 25–73 y; mean age, 40.72 ± 13.09 y). They were mainly used to analyze the changes in serum viral markers and S1P before and after treatment for the same individual. Pathological sections of liver tissues from 19 randomly selected HCC patients (14 who were HBV positive over 10 y and 5 who were HBV negative) were collected to detect HBV Ag and SphK1 by immunohistochemistry and immunofluorescence. This research was conducted by the ethical standards of the Declaration of Helsinki and was approved by the Institutional Review Board of the Fourth Affiliated Hospital of Soochow University. Informed consent was waived due to its retrospective nature, no additional sample collection, and no interference with the treatment and prognosis of patients.

C57BL/6 mice were purchased from Shanghai Slac Animal Inc. and housed in a specific-pathogen-free rodent barrier facility. All animal experiments were approved by the Institutional Animal Care and Use Committee of Soochow University. The HBV replication-competent plasmid containing 1.3 copies of the HBV genome, pUC19-HBV1.3 (pHBV1.3), HBx-depleted plasmid pHBV-ΔHBx, and plasmids encoding Myc-tagged HBs, HBx, and HBc in a backbone of pcDNA3.1 were kindly provided by Prof. Min Li (Institute of Biology and Medical Sciences, Soochow University) (19–21). USF1 cDNA was amplified from HepG2 cells by standard PCR and cloned in pEGFP-N1or pcDNA3.1 vector plasmid. The NCBI dataset was used to generate SphK1 luciferase reporter genes and cloned in pGL3 plasmid.

The human hepatoma cell line (HepG2, Huh7, HepG2-NTCP, HepG2.2.15) and human embryonic kidney cell line 293T (HEK293T) were cultured in DMEM (Life Technologies, U.S.A) or MEM (FuHeng, China), supplemented with 10% FBS (PAN, Germany), penicillin and streptomycin (100 U/ml). The cells were seeded in 24-well plates to be 80% confluent the following day. The cells were transfected with pUC19, pHBV1.3, pMyc-HBx, pMyc-ΔHBx, pMyc-HBs, and pMyc-HBc (1.0 μg each) using Lipofectamine 2000 (Thermo Fisher, 11668-019) in opti-MEM (Life Technologies) for 48 h. Negative control (si-NC) (sense: 5′-UUCUCCGAACGUGUCACGUTT-3′ and antisense: 5′-ACGUGACACGUUCGGAGAATT-3′) and USF1–small interfering RNA (siRNA) (sense: 5′-GCCGAGACAAGAUCAACAATT-3′ and antisense: 5′-UUGUUGAUCUUGUCUCGGCTT-3′) were purchased from GenePharma (China) and transfected into cells by RNATransMate (BBI, Shanghai, China) according to the instructions provided by the manufacturers.

The HBV infection experiment was performed as previously reported (22). Polyethylene glycol–concentrated HBV virion derived from HBsAg-positive patients’ serum with HBV DNA copies above 107 IU/ml, using serum from HBV-negative patients as a negative control. HepG2-NTCP cells were exposed to HBV in the presence of 2% polyethylene glycol 8000 for 24 h, which means contact with the virus. Repeated washing using PBS cleared the residual free virus. At this point, the virus had entered the cell, and this moment was marked as day 0 (D0). The cell was cultured in the DMEM supplemented with 2.5% DMSO for indicated times.

HepG2.2.15 hepatocytes were seeded in the plate by 105 cells/well. A 1.5 μg/ml concentration of entecavir (ETV) (Baraclude, China) and 5 μM FTY720 fluid (MedChemExpress, USA) were added to the culture medium, respectively, using untreated cells as control. The cells and supernatant were collected for subsequent studies after 3 d.

CRISPR/Cas9 Gene knockout kit was purchased from Suzhou Haixing Biological Technology Co. (Jiangsu, China), including one vector of gRNA sequence and specific homologous DNA fragment. The gRNA sequence is GTGAAGCGAAGTGCACACGG (23). At 3 d after viral infection of HepG2-NTCP cells, Cas9/gRNA was transfected into cells using Lipofectamine 2000, and Cas9 eliminated HBV replication under the gRNA’s lead.

The serum samples and cell culture supernatants collected from the study subjects were stored at −80°C before the measurement. S1P, Sph, dhSph, and dhS1P were measured using a high performance liquid chromatography–tandem mass spectrometry methodology. All the determinations were carried out as previously described in Central Laboratory, Xuhui Central Hospital, Shanghai, China (24). HBsAg (Abbott, USA) and HBeAg (Abbott) measured by chemiluminescence. HBV DNA was detected on a Roche LightCycler 480 (LC480) II quantitative real-time PCR (qPCR) platform using a PCR fluorescent diagnostic kit (Sansure Biotech, China) according to the manufacturer’s instructions.

Total RNA was extracted from frozen liver tissues and cultured cells using a TRIzol reagent (Thermo Fisher, 15596018) and quantified using Qubit 4 (Invitrogen). Total RNA (2 μg) was reverse transcribed, and mRNA was determined by quantitative real-time reverse transcription PCR (qRT-PCR) analysis using SYBR Green I Master (E096-01B, Novoprotein, China) and the appropriate primers (5′-GCTACGAGCTGCCTGACGG-3′ and 5′-TGTTGGCGTACAGGTCTTTGC-3′ for β-actin, 5′-GTCTGTGCCTTCTCATCTG-3′ and 5′-GTTCACGGTGGTCTCCAT-3′ for HBx, 5′-GCTCTGGTGGTCATGTCTGG-3′ and 5′-CACAGCAATAGCGTGCAGT-3′ for SphK1, and 5′-ATCGTGCAGCTCTCCAAGATAATCC-3′ and 5′-AGTTCTTCAGACAAGCGGTGGTTAC-3′ for USF1). The primers were synthesized by Sangon Biotech (Shanghai, China) Company. Relative changes in gene expression levels were determined using the 2-ΔΔct method. The cycle number at which the transcripts were detectable (Ct) was normalized to the cycle number of β-Actin gene detection, referred to as ΔCt.

The total cells were lysed in radioimmunoprecipitation assay buffer (Beyotime Biotechnology, China). The protein concentrations of the whole cells were determined by the BCA (Beyotime Biotechnology) method using the Beyotime protein assay kit. Cell lysate was separated in a 10% polyacrylamide gel and transferred to a nitrocellulose membrane. The membrane was blocked with 5% skim milk in PBST for 1 h and then incubated at 4°C overnight with anti–β-ACTIN (Proteintech, USA, 66009-1-Ig) and anti-SphK1 (Proteintech, 10670-1-AP). The membranes were washed with PBST and incubated for 1 h with HRP-conjugated secondary Ab (Proteintech, SA00001-2).

HEK293T cells were cotransfected using Lipofectamine 2000 transfection reagent (Thermo Fisher, 11668-019) with 0.1 μg of luciferase reporter plasmids pGL3-SphK1, and 0.1 μg pcDNA3.1-USF1 or its empty vector (pcDNA3.1) individually and 50 ng thymidine kinase promoter–Renilla luciferase reporter plasmid (transfection control) as described previously (25). 48 h later, the cell lysates were prepared, and the luciferase activities were determined by the dual-luciferase reporter assay system (Vazyme, DD1205-01) according to the manufacturer’s instructions.

The 6-wk-old mice were injected within 10 s via the tail vein with plasmid DNA diluted in PBS to a volume equivalent to 10% of the total body weight of each animal. Each mouse received 10 μg of pHBV1.3 plasmid or its vector control plasmid pUC19. Mice serum and liver samples were collected at the indicated times.

Chromatin immunoprecipitation (ChIP) assay was performed using the ChIP kit (CST, USA) following the manufacturer’s protocol. Briefly, HepG2 was cross-linked using 1% formaldehyde and then treated with glycine. Subsequently, ultrasonication was performed to break the chromatin. The chromatin fragments were precipitated with anti-IgG or anti-USF1 at 4°C. After treatment with NaCl and protease K, the DNA was purified, and the enrichment of SphK1 was measured by PCR.

Statistical significance for all experiments was determined by using Student t test or the Mann–Whitney rank sum test (GraphPad Prism 5 software). Error bars are reported as standard errors of the means, and significance was assigned for p values of <0.05.

In this study, 20 patients with CHB received direct anti-HBV therapy using NAs, which effectively blocked viral replication, significantly reducing HBV DNA copies in serum (Fig. 1A). Despite this, virus components (HBsAg and HBeAg) and transaminases (alanine aminotransferase and aspartate aminotransferase) in serum showed no significant changes (Fig. 1B–E), consistent with previous reports that ETV could profoundly suppress HBV DNA but rarely achieve HBsAg loss in the short term (26). Interestingly, levels of SphK-associated sphingolipid metabolites, including Sph, S1P, dhSph, and dhS1P, were evaluated, and only S1P and Sph were significantly reduced after antiviral treatment (Fig. 1F–I). A positive correlation between HBV DNA and serum S1P levels was observed (r = 0.4357, p = 0.0049) (Fig. 1J).

FIGURE 1.

Serum S1P level was decreased in CHB patients treated with anti-HBV drug NAs. Blood samples were collected from 20 cases of pre- and post-treatment hepatitis B patients undergoing anti-HBV treatment by nucleos(t)ide analogs (NAs) for at least 3 months. (A) HBV DNA was detected by qPCR, 100 IU/ml of the minimum detection limit. (B) HBsAg was detected by chemiluminescence, positive above 0.05 IU/ml. (C) HBeAg was detected by chemiluminescence, positive above 1 S/CO. (D and E) Liver function alanine aminotransferase and aspartate aminotransferase by chemiluminescent assay. (F–I) S1P and related sphingolipid metabolites were detected by mass spectrometry. (J) Correlation between HBV DNA and serum S1P. The differences between pre- and post-treatment groups were analyzed by paired Student t test. The statistical significance was defined as p ≤ 0.05.

FIGURE 1.

Serum S1P level was decreased in CHB patients treated with anti-HBV drug NAs. Blood samples were collected from 20 cases of pre- and post-treatment hepatitis B patients undergoing anti-HBV treatment by nucleos(t)ide analogs (NAs) for at least 3 months. (A) HBV DNA was detected by qPCR, 100 IU/ml of the minimum detection limit. (B) HBsAg was detected by chemiluminescence, positive above 0.05 IU/ml. (C) HBeAg was detected by chemiluminescence, positive above 1 S/CO. (D and E) Liver function alanine aminotransferase and aspartate aminotransferase by chemiluminescent assay. (F–I) S1P and related sphingolipid metabolites were detected by mass spectrometry. (J) Correlation between HBV DNA and serum S1P. The differences between pre- and post-treatment groups were analyzed by paired Student t test. The statistical significance was defined as p ≤ 0.05.

Close modal

Hepatic tissues of randomly selected 19 patients, including 14 primary hepatitis B–related HCC and 5 HBV-negative HCC, were retrospectively analyzed to investigate SphK1 expression (Table I). Immunohistochemistry revealed varying degrees of HBsAg positivity, ranging from weakly positive (±) to strongly positive (1+ to 4+), with an uneven distribution and striking heterogeneity. SphK1 was commonly expressed in the liver tissues of all 19 patients, with localized high expression in a lamellar form. Notably, four patients exhibited significantly enhanced SphK1 expression in HBsAg-positive regions. This enhanced expression is illustrated in Fig. 2 (A and B) for patient 1 and in supplementary data for patients 2–4 (Supplemental Fig. 1). Furthermore, immunofluorescence staining confirmed the coexpression of SphK1 and HBsAg in the cytoplasm of the same hepatocyte (Fig. 2C, 2D). These findings indicate that HBV-colonized liver tissues are a significant source of high SphK1 expression in some HBV-related HCC patients.

Table I.
Detailed information of 19 HCC patients
No.SexAge (y)Classification and Differentiation of Liver CancerHistory of HBV (y)HBsAg (IU/ml)Immunohistochemistry Resultsa
HBsAgSphK1Coexpressed Sites of HBsAg and SphK1
Male 44 Medium-low differentiated HCC 20 >250 ++++
inhomogeneous 
++++
inhomogeneous 
++++ 
Female 52 Medium differentiated HCC 20 >250 ++++
inhomogeneous 
++++
inhomogeneous 
++++ 
Male 51 High differentiated HCC 10 >250 ++ 
Male 52 Medium-low differentiated HCC 20 68.49 ++ +++ ++ 
Female 74 Medium-low differentiated HCC 15 883.71 ++ − 
Male 51 Medium differentiated HCC 10 >250 ++ +++ − 
Female 51 Medium-low differentiated HCC 30 >250 ± ++++ − 
Male 52 Medium differentiated HCC 20 >250 ++ − 
Male 45 Low differentiated HCC 10 >250 ± − 
10 Male 52 Medium-low differentiated HCC 15 101.87 − − 
11 Female 51 Medium-low differentiated HCC 30 >250 ± +++ − 
12 Female 54 Low differentiated HCC 20 108.54 ++ − 
13 Male 40 Medium-low differentiated HCC 10 >250 ± ++ − 
14 Female 62 Medium differentiated HCC 10 >250 ++ − 
15 Male 72 Negative control − − 
16 Male 79 Negative control − ++++ − 
17 Male 56 Negative control − ++ − 
18 Female 50 Negative control 0.01 −  
19 Female 74 Negative control − − 
No.SexAge (y)Classification and Differentiation of Liver CancerHistory of HBV (y)HBsAg (IU/ml)Immunohistochemistry Resultsa
HBsAgSphK1Coexpressed Sites of HBsAg and SphK1
Male 44 Medium-low differentiated HCC 20 >250 ++++
inhomogeneous 
++++
inhomogeneous 
++++ 
Female 52 Medium differentiated HCC 20 >250 ++++
inhomogeneous 
++++
inhomogeneous 
++++ 
Male 51 High differentiated HCC 10 >250 ++ 
Male 52 Medium-low differentiated HCC 20 68.49 ++ +++ ++ 
Female 74 Medium-low differentiated HCC 15 883.71 ++ − 
Male 51 Medium differentiated HCC 10 >250 ++ +++ − 
Female 51 Medium-low differentiated HCC 30 >250 ± ++++ − 
Male 52 Medium differentiated HCC 20 >250 ++ − 
Male 45 Low differentiated HCC 10 >250 ± − 
10 Male 52 Medium-low differentiated HCC 15 101.87 − − 
11 Female 51 Medium-low differentiated HCC 30 >250 ± +++ − 
12 Female 54 Low differentiated HCC 20 108.54 ++ − 
13 Male 40 Medium-low differentiated HCC 10 >250 ± ++ − 
14 Female 62 Medium differentiated HCC 10 >250 ++ − 
15 Male 72 Negative control − − 
16 Male 79 Negative control − ++++ − 
17 Male 56 Negative control − ++ − 
18 Female 50 Negative control 0.01 −  
19 Female 74 Negative control − − 
a

±, weakly positive defined as several brown dots < 5 on the entire field; 1+ to 4+, positive intensity from linear to patchy regions on the entire field.

FIGURE 2.

Hyperexpression of SphK1 in HBV-positive HCC patients by liver biopsy tissue analysis. HBsAg and SphK1 were determined by immunohistochemistry analysis in liver tissues from primary HCC patients owing to chronic HBV infection. Coexpressed HBsAg and SphK1 were detected by immunofluorescence. The results were shown from patient 1 with over 250 IU/ml serum S1P. (A) HBsAg and SphK1 were presented in the whole tissue section with primary magnification. Scale bars, 4 mm. (B) HBsAg and SphK1 in the same tissue site were shown in magnification (×200). Scale bars, 200 μm. Brown represents protein expression. HBsAg is labeled in red, SphK1 is labeled in green, and merged is labeled in orange. (C and D) Primary magnification for (C) and 200× magnification for (D). Scale bars, 5 mm (C) and 100 μm (D).

FIGURE 2.

Hyperexpression of SphK1 in HBV-positive HCC patients by liver biopsy tissue analysis. HBsAg and SphK1 were determined by immunohistochemistry analysis in liver tissues from primary HCC patients owing to chronic HBV infection. Coexpressed HBsAg and SphK1 were detected by immunofluorescence. The results were shown from patient 1 with over 250 IU/ml serum S1P. (A) HBsAg and SphK1 were presented in the whole tissue section with primary magnification. Scale bars, 4 mm. (B) HBsAg and SphK1 in the same tissue site were shown in magnification (×200). Scale bars, 200 μm. Brown represents protein expression. HBsAg is labeled in red, SphK1 is labeled in green, and merged is labeled in orange. (C and D) Primary magnification for (C) and 200× magnification for (D). Scale bars, 5 mm (C) and 100 μm (D).

Close modal

To validate the correlation between HBV and SphK1, we established an HBV infection model in HepG2 cells overexpressing the HBV receptor NTCP (HepG2-NTCP) (19). After exposing HepG2-NTCP cells to HBV virions for 24 h, we repeatedly washed the cells with PBS to move residual virus, making this as day 0 (D0). HBV serum biomarkers (HBsAg, HBeAg, and HBV DNA) increased with HBV infection, peaking 3 days postinfection (Fig. 3A–C). Compared to uninfected controls, intracellular SphK1 and released S1P levels were strongly correlated with HBV replication during persistent infection (Fig. 3D–F). Interestingly, SphK1 expression decreased on day 0, likely due to compromised membrane integrity during massive viral entry. Because S1P is the final product of sphingolipid metabolism, and sphingolipids are ubiquitous components of mammalian membranes, this observation aligns with their role in cellular processes (27). These results suggest that HBV regulates SphK1 expression and S1P generation once the virus establishes a stable presence within the cell.

FIGURE 3.

SphK1–S1P expression is significantly increased in HBV-infected HepG2-NTCP cells. HepG2-NTCP cells (105/well) were incubated with HBV virion derived from HBsAg-positive patients’ serum with HBV DNA copies above 107 IU/ml for 24 h, followed by repeated washing with PBS. At this point, the virus had entered the cell, and this moment was marked as day 0 (D0). Supernatants were collected at days 0, 3, 6, and 9. (A–C) Relative HBsAg and HBeAg levels in the supernatant were measured by chemiluminescence (A and B), and HBV DNA copies (C) were detected by qPCR. (D and E) Extracellular S1P and intracellular SphK1 mRNA were detected by mass spectrometry and qRT-PCR. (F) Western blot analysis of SphK1 expression level from HBV-infected HepG2-NTCP cells using anti-SphK1 Ab and β-ACTIN was used as a loading control. The data shown are representative of three independent experiments (n = 6) with similar results, and significance was determined by Student t test. The statistical significance level was defined as p ≤ 0.05.

FIGURE 3.

SphK1–S1P expression is significantly increased in HBV-infected HepG2-NTCP cells. HepG2-NTCP cells (105/well) were incubated with HBV virion derived from HBsAg-positive patients’ serum with HBV DNA copies above 107 IU/ml for 24 h, followed by repeated washing with PBS. At this point, the virus had entered the cell, and this moment was marked as day 0 (D0). Supernatants were collected at days 0, 3, 6, and 9. (A–C) Relative HBsAg and HBeAg levels in the supernatant were measured by chemiluminescence (A and B), and HBV DNA copies (C) were detected by qPCR. (D and E) Extracellular S1P and intracellular SphK1 mRNA were detected by mass spectrometry and qRT-PCR. (F) Western blot analysis of SphK1 expression level from HBV-infected HepG2-NTCP cells using anti-SphK1 Ab and β-ACTIN was used as a loading control. The data shown are representative of three independent experiments (n = 6) with similar results, and significance was determined by Student t test. The statistical significance level was defined as p ≤ 0.05.

Close modal

To further clarify the HBV-dependent alterations, we treated HBV-replicating HepG2.2.15 cells with antiviral agents. Upon suppression of HBV reproduction by the direct antiviral agent entecavir, intracellular SphK1, and extracellular S1P levels significantly decreased (Fig. 4A–C). Additionally, we used the CRISPR-Cas9 system (HBV-gRNA/Cas9) to cleave the HBV genome (28, 29). As shown in Fig. 4D, the plasmids (gRNA/Cas9) successfully reduced the HBV genome, which was accompanied by a significant reduction in viral protein Ag. As expected, the inhibition of HBV replication also significantly diminished SphK1 expression and the release of extracellular S1P (Fig. 4E, 4F). Given the enhancement of SphK1–S1P with the HBV infection, we further investigated its effects on HBV replication. We treated HepG2.2.15 cells with FTY720, a competitive analog of S1P, which blocked the SphK1–S1P–S1PRs axis and inhibited HBV DNA replication (Fig. 4A–C). However, merely knocking down SphK1 expression via siRNA did not significantly inhibit HBV replication (Supplemental Fig. 2), consistent with other reports (30, 31). These data indicate that the SphK1–S1P–S1PRs axis may influence HBV replication by more complex mechanisms.

FIGURE 4.

Inhibition of HBV reduces the SphK1 and S1P generations. HepG2.2.15 cells (105/well) were treated with entecavir (ETV) and SphK1 selective inhibitor (FTY720) for 3 d. CRISPR-Cas9 system (gRNA) or its vector control (eGFP-NC) were transfected into HBV-infected HepG2-NTCP cells to cleavage the HBV genome. (A and D) Supernatants were collected and subjected to determine HBsAg and HBeAg levels by chemiluminescence, and HBV DNA copies by qPCR. (B and E) Extracellular S1P and intracellular SphK1 mRNA were detected by mass spectrometry and qRT-PCR. (C and F) SphK1 protein level was detected by Western blot, and β-ACTIN was used as a loading control. The data shown are representative of three independent experiments (n = 4) with similar results, and significance was determined by Student t test. The statistical significance level was defined as p ≤ 0.05.

FIGURE 4.

Inhibition of HBV reduces the SphK1 and S1P generations. HepG2.2.15 cells (105/well) were treated with entecavir (ETV) and SphK1 selective inhibitor (FTY720) for 3 d. CRISPR-Cas9 system (gRNA) or its vector control (eGFP-NC) were transfected into HBV-infected HepG2-NTCP cells to cleavage the HBV genome. (A and D) Supernatants were collected and subjected to determine HBsAg and HBeAg levels by chemiluminescence, and HBV DNA copies by qPCR. (B and E) Extracellular S1P and intracellular SphK1 mRNA were detected by mass spectrometry and qRT-PCR. (C and F) SphK1 protein level was detected by Western blot, and β-ACTIN was used as a loading control. The data shown are representative of three independent experiments (n = 4) with similar results, and significance was determined by Student t test. The statistical significance level was defined as p ≤ 0.05.

Close modal

The enhanced availability of promoter-bound TFs is crucial for upregulating promoter activity and gene expression. In HBV infection, the upregulation of SphK1 at the mRNA level suggests that it might be regulated by transcription factors (TFs). To identify the TFs regulating SphK1 expression during HBV infection, we analyzed publicly available ChIP sequencing datasets. We identified 118 TFs and visualized the top 15 based on their binding score (Fig. 5A). We then examined the expression of these 15 TFs in HBV plasmid-transfected Huh7 cells and HBV-infected HepG2-NTCP cells using qRT-PCR. Notably, HBV infection caused a substantial increase in USF1 mRNA compared with control cells, which corresponded with high SphK1 expression (Fig. 5B, 5C). Further examination of USF1 expression in HepG2-NTCP cells revealed a continuous increase in USF1 levels with persistent HBV infection, mirroring the trend in SphK1 expression (Fig. 5D). Additionally, transfecting Huh7 cells with different HBV ORF plasmids showed that SphK1 expression notably increased only with the replication-competent HBV plasmid pHBV1.3. As expected, the trend of USF1 expression was consistent with that of SphK1 (Fig. 5E). Taken together, these data suggest that the transcription factor USF1 plays a significant role in HBV-mediated enhancement of the SphK1–S1P pathway.

FIGURE 5.

HBV-mediated alteration in the expression of transcription factors binding to SphK1 promoter. The publicly available ChIP sequencing dataset (Signaling Pathways Project) was used to forecast the transcription factors that bind to the SphK1 promoter. (A) The top 15 transcription factors that bind to the SphK1 were visualized according to the descending order of binding score. (B and C) qRT-PCR analysis measured the top 15 transcription factors mRNA level, and high expression of USF1 was screened out. HepG2-NTCP cells (105/well) were incubated with HBV virion derived from HBsAg-positive patients serum with HBV DNA copies above 107 IU/ml for 24 h, followed by repeated washing with PBS. (D) Supernatants were collected at days 0, 3, 6, and 9, followed by detection of USF1 expression level. (E) Huh7 cells (105/well) were cotransfected with HBV, HBx, ΔHBx, HBsAg, HBcAg, and its vector control plasmids for 48 h, followed by detection of SphK1 and USF1 expression level. The data shown are representative of three independent experiments (n = 4) with similar results, and significance was determined by Student t test. The statistical significance level was defined as p ≤ 0.05.

FIGURE 5.

HBV-mediated alteration in the expression of transcription factors binding to SphK1 promoter. The publicly available ChIP sequencing dataset (Signaling Pathways Project) was used to forecast the transcription factors that bind to the SphK1 promoter. (A) The top 15 transcription factors that bind to the SphK1 were visualized according to the descending order of binding score. (B and C) qRT-PCR analysis measured the top 15 transcription factors mRNA level, and high expression of USF1 was screened out. HepG2-NTCP cells (105/well) were incubated with HBV virion derived from HBsAg-positive patients serum with HBV DNA copies above 107 IU/ml for 24 h, followed by repeated washing with PBS. (D) Supernatants were collected at days 0, 3, 6, and 9, followed by detection of USF1 expression level. (E) Huh7 cells (105/well) were cotransfected with HBV, HBx, ΔHBx, HBsAg, HBcAg, and its vector control plasmids for 48 h, followed by detection of SphK1 and USF1 expression level. The data shown are representative of three independent experiments (n = 4) with similar results, and significance was determined by Student t test. The statistical significance level was defined as p ≤ 0.05.

Close modal

To investigate the regulatory effect of USF1 on SphK1 transcription, we constructed a luciferase reporter plasmid encoding the full-length SphK1 promoter and assessed whether USF1 regulates SphK1 transcription. HEK293T cells were cotransfected with USF1 plasmids and the SphK1 reporter plasmids, and fluorescence microscopy showed that USF1 localized in the nucleus (Fig. 6A). The luciferase reporter gene assay revealed that USF1 significantly enhanced the transcriptional activity of the SphK1 promoter (Fig. 6B). To determine whether USF1 could directly bind to the SphK1 promoter, we performed a ChIP assay. The results showed deeper binds for SphK1 promoter in the USF1 group compared with the IgG isotype control, indicating that the USF1 binds to the SphK1 promoter (Fig. 6C). To further elucidate USF1’s regulatory effect on SphK1 during HBV infection, we examined SphK1 expression following knockdown of endogenous USF1 in HBV-infected HepG2-NTCP and Huh7 cells. siRNA-mediated knockdown of USF1 significantly reduced USF1 expression levels (Fig. 6D, 6F) and resulted in a significant decrease in SphK1 expression compared with the si-NC group in both cell models (Fig. 6E, 6G). These results demonstrate that HBV infection triggers nuclear transfer and expression of USF1, which in turn activates the SphK1 promoter.

FIGURE 6.

HBV activates the SphK1 promoter through the activation of USF1. HEK239T cells were transfected with pEGFP-USF1 for 48 h. The cells were then fixed, penetrated, and subjected to staining with DAPI. (A) Representative composite fluorescence microscopy images showed the location of USF1 (green) and nuclear (blue, DAPI) at 200× magnification. (B) Luciferase activity in HEK293T cells transfected for 48 h with constructs expressing an SphK1–luciferase reporter together with a USF1-expressing plasmid. (C) HepG2 cells were detected by SphK1 promoter primers after immunoprecipitation experiments using IgG and USF1 Abs, respectively. (D and F) si-USF1 or si-NC (negative control) were transfected into HBV-infected HepG2-NTCP cells or HBV plasmid-transfected Huh7 cells. (E and G) The knockdown efficiency of USF1 was assessed by qRT-PCR. SphK1 mRNA and protein levels were evaluated by qRT-PCR or Western blot analysis. The data shown are representative of three independent experiments (n = 4) with similar results, and significance was determined by Student t test. The statistical significance level was defined as p ≤ 0.05.

FIGURE 6.

HBV activates the SphK1 promoter through the activation of USF1. HEK239T cells were transfected with pEGFP-USF1 for 48 h. The cells were then fixed, penetrated, and subjected to staining with DAPI. (A) Representative composite fluorescence microscopy images showed the location of USF1 (green) and nuclear (blue, DAPI) at 200× magnification. (B) Luciferase activity in HEK293T cells transfected for 48 h with constructs expressing an SphK1–luciferase reporter together with a USF1-expressing plasmid. (C) HepG2 cells were detected by SphK1 promoter primers after immunoprecipitation experiments using IgG and USF1 Abs, respectively. (D and F) si-USF1 or si-NC (negative control) were transfected into HBV-infected HepG2-NTCP cells or HBV plasmid-transfected Huh7 cells. (E and G) The knockdown efficiency of USF1 was assessed by qRT-PCR. SphK1 mRNA and protein levels were evaluated by qRT-PCR or Western blot analysis. The data shown are representative of three independent experiments (n = 4) with similar results, and significance was determined by Student t test. The statistical significance level was defined as p ≤ 0.05.

Close modal

To investigate the effect of HBV on SphK1–S1P in vivo, 10 ug of pUC19-HBV1.3 plasmid was injected into mice by high-pressure tail vein. Serum and liver samples were collected from the mice on day 4. Serum testing revealed a significant enhancement of viral replication along with increased S1P expression (Fig. 7A). Liver assays showed that HBV replication led to increased expression of USF1 and SphK1 (Fig. 7B). Additionally, liver samples were collected on days 1, 2, 3, and 4. The results showed that HBV replication (indicated by HBx expression levels) gradually increased over time, with significant increases in USF1 and SphK1 expression on day 4 (Fig. 7C).

FIGURE 7.

Exposure to HBV in vivo promotes the expression of USF1 and SphK1–S1P. A total of 10 μg of pUC19 and pUC19-HBV1.3(pHBV1.3) were injected into C57BL/6 mice through a hydrodynamic injection, and the mice were sacrificed on days 0, 1, 2, 3, and 4. (A) The mouse serum on day 4 was collected to measure the level of HBsAg and HBeAg by chemiluminescence and the level of S1P by mass spectrometry. (B and C) The liver tissue RNA was extracted to measure the level of HBx, USF1, and SphK1 by qRT-PCR. The data shown are representative of three independent experiments (n = 6) with similar results, and significance was determined by Student t test. The statistical significance level was defined as p ≤ 0.05.

FIGURE 7.

Exposure to HBV in vivo promotes the expression of USF1 and SphK1–S1P. A total of 10 μg of pUC19 and pUC19-HBV1.3(pHBV1.3) were injected into C57BL/6 mice through a hydrodynamic injection, and the mice were sacrificed on days 0, 1, 2, 3, and 4. (A) The mouse serum on day 4 was collected to measure the level of HBsAg and HBeAg by chemiluminescence and the level of S1P by mass spectrometry. (B and C) The liver tissue RNA was extracted to measure the level of HBx, USF1, and SphK1 by qRT-PCR. The data shown are representative of three independent experiments (n = 6) with similar results, and significance was determined by Student t test. The statistical significance level was defined as p ≤ 0.05.

Close modal

The SphK1–S1P axis has been recognized for its critical role in hepatic inflammatory injury, liver fibrosis, and HCC. S1P released from the liver in response to injury promotes the recruitment of various immune cells, leading to hepatic inflammation (32). Additionally, S1P regulates signaling and physiological processes across several pathways to promote HCC (16). Given the importance of SphK1–S1P in liver injury, this study focused on whether HBV could directly alter SphK1–S1P expression. Although SphK1–S1P–S1PRs signaling has been reported to influence viral activity and host defense immunity, few studies have explored the mechanisms by which the virus alters sphingolipid metabolism (2, 33)

Our findings reveal a relationship between HBV and SphK1–S1P in serum and liver tissue samples from patients. Retrospective analysis of patients treated with NAs for over 3 months showed a positive correlation between HBV DNA and S1P levels. SphK1 hyperexpression predominantly colocalized in HBV-positive regions of liver tissues from some HBV-positive HCC patients. These results suggest a connection between HBV and SphK1 in the process of HBV-related liver immune injury and tumorigenesis. In vitro, HBV-dependent SphK1 expression was observed at both the mRNA and protein levels, indicating that HBV infection can upregulate SphK1–S1P production. Serum S1P has the potential to be serve as a unique indicator for monitoring sphingolipid metabolism in HBV-infected hepatocytes. Current biomarkers for HBV replication mainly consist of the viral components or nucleic acids, which do not reflect the metabolic status of infected cells.

HBV infection induced upregulation of SphK1 at the mRNA level and increased activity, as indicated by elevated S1P, suggesting an association with TFs. Bioinformatic analysis of ChIP sequencing data indicated that several TFs might be involved in regulating SphK1 promoters. Notably, USF1 was verified as the strongest TF for SphK1 transcription according to our data. Although the role of USF1 in liver diseases is well established, its response to HBV infection remains controversial. Chen et al. (34) and Zeng et al. (35) reported that USF1 mRNA expression was increased in HCC lines compared with the normal liver cell lines and was not associated with HBsAg status. However, Baidya et al. (36) found that HBx-induced hypermethylation of the USF1 promoter led to the downregulation of USF1. AP2α is another TF related to target SphK1 transcription in response HBx (3). In our study, the complete replication activity of HBV, rather than individual viral components, appeared to be the key factor in enhancing USF1 activity, partially explaining these inconsistent reports. Overall, HBV infection manipulates sphingolipid metabolism by regulating potential TFs, which may respond to viral activity and viral components.

The SphK1–S1P–S1PRs axis influences various biological functions. S1P interacts with extracellular receptors to modulate downstream signals and acts as a secondary messenger directly involved in intracellular signal transduction and regulation (13). S1P modular, as active phospholipid-related drugs, have the potential to balance immune cell trafficking and activation, affect the cell cycle, inhibit viral replication, and perform other biological functions. For example, FTY720, a prodrug phosphorylated to an S1P analog, has been approved by U.S. Food and Drug Administration for the treatment of multiple sclerosis and cardio protection (37). Our results indicated that FTY720 not only inhibited the SphK1 expression but also interfered with HBV replication. Considering that simply knocking down SphK1 expression did not affect HBV replication, this suggests that FTY720 may interfere with viral activity through other complex pathways, such as acting on different S1PRs and relying on some positive/negative feedback mechanisms of S1P in different cells (38).

In summary, we discovered that HBV infection directly manipulated USF1 to enhance SphK1 transcription and increase S1P production. These underlying molecular mechanisms expand our understanding of the interplay between hepatitis B and sphingolipid metabolism. The potency of these pathways has led to the development of drugs targeting SphK1–S1P–S1PR signaling. HBV-mediated activation of SphK1–S1P may serve as a novel surveillance indicator for HBV-associated liver injury and provide new therapeutic targets for delaying HBV-associated liver injury, liver fibrosis, cirrhosis, and hepatocellular carcinoma.

The authors have no financial conflicts of interest.

This work was supported by Suzhou Science and Technology Planning Project Grants SLT201921, SZM2021011, SZM2021018, and SKY2022091.

The online version of this article contains supplemental material.

CHB

chronic hepatitis B

ChIP

chromatin immunoprecipitation

dhS1P

dihydro-S1P

dhSph

dihydrosphingosine

ETV

entecavir

FTY720

fingolimod

gRNA

guide RNA

HBeAg

hepatitis B e antigen

HBsAg

hepatitis B surface antigen

HBV

hepatitis B virus

HCC

hepatocellular carcinoma

NA

nucleos(t)ide analog

NTCP

sodium taurocholate cotransporting polypeptide

ORF

open reading frame

pHBV1.3

pUC19-HBV1.3

qPCR

quantitative real-time PCR

qRT-PCR

quantitative real-time reverse transcription PCR

S1P

sphingosine-1-phosphate

siRNA

small interfering RNA

Sph

sphingosine

SphK

sphingosine kinase

TF

transcription factor

1
Sarmati
,
L.
,
V.
Malagnino
.
2019
.
HBV infection in HIV-driven immune suppression
.
Viruses
11
:
1077
.
2
Zhang
,
L.
,
J.
Liu
,
E.
Xiao
,
Q.
Han
,
L.
Wang
.
2023
.
Sphingosine-1-phosphate related signalling pathways manipulating virus replication
.
Rev. Med. Virol.
33
:
e2415
.
3
Lu
,
Z. P.
,
Z. L.
Xiao
,
Z.
Yang
,
J.
Li
,
G. X.
Feng
,
F. Q.
Chen
,
Y. H.
Li
,
J. Y.
Feng
,
Y. E.
Gao
,
L. H.
Ye
,
X. D.
Zhang
.
2015
.
Hepatitis B virus X protein promotes human hepatoma cell growth via upregulation of transcription factor AP2α and sphingosine kinase 1
.
Acta Pharmacol. Sin.
36
:
1228
1236
.
4
Maini
,
M. K.
,
A.
Schurich
.
2010
.
The molecular basis of the failed immune response in chronic HBV: therapeutic implications
.
J. Hepatol.
52
:
616
619
.
5
Chua
,
C.
,
D.
Mahamed
,
S.
Nkongolo
,
J. D.
Sanchez Vasquez
,
A.
Mehrotra
,
D. K. H.
Wong
,
R. T.
Chung
,
J. J.
Feld
,
H. L. A.
Janssen
,
A. J.
Gehring
.
2024
.
Liver-restricted type I IFN signature precedes liver damage in chronic hepatitis B patients stopping antiviral therapy
.
J. Immunol.
212
:
1002
1011
.
6
Resop
,
R. S.
,
A.
Bosque
.
2022
.
Pharmacological targeting of sphingosine kinases impedes HIV-1 infection of CD4 T cells through SAMHD1 modulation
.
J. Virol.
96
:
e0009622
.
7
Shu
,
X.
,
W.
Wu
,
R. D.
Mosteller
,
D.
Broek
.
2002
.
Sphingosine kinase mediates vascular endothelial growth factor-induced activation of ras and mitogen-activated protein kinases
.
Mol. Cell. Biol.
22
:
7758
7768
.
8
Dutton
,
E. E.
,
D. W.
Gajdasik
,
C.
Willis
,
R.
Fiancette
,
E. L.
Bishop
,
A.
Camelo
,
M. A.
Sleeman
,
M.
Coccia
,
A. M.
Didierlaurent
,
M.
Tomura
, et al
.
2019
.
Peripheral lymph nodes contain migratory and resident innate lymphoid cell populations
.
Sci. Immunol.
4
:
eaau8082
.
9
Snider
,
A. J.
,
K. A.
Orr Gandy
,
L. M.
Obeid
.
2010
.
Sphingosine kinase: role in regulation of bioactive sphingolipid mediators in inflammation
.
Biochimie
92
:
707
715
.
10
Maceyka
,
M.
,
H.
Sankala
,
N. C.
Hait
,
H.
Le Stunff
,
H.
Liu
,
R.
Toman
,
C.
Collier
,
M.
Zhang
,
L. S.
Satin
,
A. H.
Merrill
, Jr.
, et al
.
2005
.
SphK1 and SphK2, sphingosine kinase isoenzymes with opposing functions in sphingolipid metabolism
.
J. Biol. Chem.
280
:
37118
37129
.
11
Zeng
,
M.
,
A.
Feng
,
L.
Wang
,
K.
Li
,
J.
Zhou
.
2023
.
Aralia saponin A isolated from Achyranthes bidentata Bl. ameliorates LPS/D-GalN induced acute liver injury via SPHK1/S1P/S1PR1 pathway in vivo and in vitro
.
Int. Immunopharmacol.
124
:
110912
.
12
Lan
,
T.
,
C.
Li
,
G.
Yang
,
Y.
Sun
,
L.
Zhuang
,
Y.
Ou
,
H.
Li
,
G.
Wang
,
T.
Kisseleva
,
D.
Brenner
,
J.
Guo
.
2018
.
Sphingosine kinase 1 promotes liver fibrosis by preventing miR-19b-3p–mediated inhibition of CCR2
.
Hepatology
68
:
1070
1086
.
13
Li
,
L.
,
H.
Wang
,
J.
Zhang
,
Y.
Sha
,
F.
Wu
,
S.
Wen
,
L.
He
,
L.
Sheng
,
Q.
You
,
M.
Shi
, et al
.
2020
.
SPHK1 deficiency protects mice from acetaminophen-induced ER stress and mitochondrial permeability transition
.
Cell Death Differ.
27
:
1924
1937
.
14
Avni
,
D.
,
K. B.
Harikumar
,
A. J.
Sanyal
,
S.
Spiegel
.
2021
.
Deletion or inhibition of SphK1 mitigates fulminant hepatic failure by suppressing TNFα-dependent inflammation and apoptosis
.
FASEB J.
35
:
e21415
.
15
Brinkmann
,
V.
,
A.
Billich
,
T.
Baumruker
,
P.
Heining
,
R.
Schmouder
,
G.
Francis
,
S.
Aradhye
,
P.
Burtin
.
2010
.
Fingolimod (FTY720): discovery and development of an oral drug to treat multiple sclerosis
.
Nat. Rev. Drug Discov.
9
:
883
897
.
16
Maceyka
,
M.
,
T.
Rohrbach
,
S.
Milstien
,
S.
Spiegel
.
2020
.
Role of sphingosine kinase 1 and sphingosine-1-phosphate axis in hepatocellular carcinoma
.
Handb. Exp. Pharmacol.
259
:
3
17
.
17
Wang
,
E.
,
X.
He
,
M.
Zeng
.
2018
.
The role of S1P and the related signaling pathway in the development of tissue fibrosis
.
Front. Pharmacol.
9
:
1504
.
18
Terrault
,
N. A.
,
A. S. F.
Lok
,
B. J.
McMahon
,
K. M.
Chang
,
J. P.
Hwang
,
M. M.
Jonas
,
R. S.
Brown
, Jr.
,
N. H.
Bzowej
,
J. B.
Wong
.
2018
.
Update on prevention, diagnosis, and treatment of chronic hepatitis B: AASLD 2018 hepatitis B guidance
.
Hepatology
67
:
1560
1599
.
19
Song
,
Y.
,
M.
Li
,
Y.
Wang
,
H.
Zhang
,
L.
Wei
,
W.
Xu
.
2021
.
E3 ubiquitin ligase TRIM21 restricts hepatitis B virus replication by targeting HBx for proteasomal degradation
.
Antiviral Res.
192
:
105107
.
20
Xu
,
J.
,
X.
Yun
,
J.
Jiang
,
Y.
Wei
,
Y.
Wu
,
W.
Zhang
,
Y.
Liu
,
W.
Wang
,
Y.
Wen
,
J.
Gu
.
2010
.
Hepatitis B virus X protein blunts senescence-like growth arrest of human hepatocellular carcinoma by reducing Notch1 cleavage
.
Hepatology
52
:
142
154
.
21
Zhong
,
L.
,
W.
Shu
,
W.
Dai
,
B.
Gao
,
S.
Xiong
.
2017
.
Reactive oxygen species-mediated c-Jun NH2-terminal kinase activation contributes to hepatitis B virus X protein-induced autophagy via regulation of the Beclin-1/Bcl-2 interaction
.
J. Virol.
91
:
e00001-17
.
22
Yang
,
Y.
,
X.
Zhao
,
Z.
Wang
,
W.
Shu
,
L.
Li
,
Y.
Li
,
Z.
Guo
,
B.
Gao
,
S.
Xiong
.
2020
.
Nuclear sensor interferon-inducible protein 16 inhibits the function of hepatitis B virus covalently closed circular DNA by integrating innate immune activation and epigenetic suppression
.
Hepatology
71
:
1154
1169
.
23
Guo
,
X.
,
P.
Chen
,
X.
Hou
,
W.
Xu
,
D.
Wang
,
T. Y.
Wang
,
L.
Zhang
,
G.
Zheng
,
Z. L.
Gao
,
C. Y.
He
, et al
.
2016
.
The recombined cccDNA produced using minicircle technology mimicked HBV genome in structure and function closely
.
Sci. Rep.
6
:
25552
.
24
Chen
,
Q.
,
W.
Wang
,
M. F.
Xia
,
Y. L.
Lu
,
H.
Bian
,
C.
Yu
,
X. Y.
Li
,
M. A.
Vadas
,
X.
Gao
,
H. D.
Lin
,
P.
Xia
.
2021
.
Identification of circulating sphingosine kinase-related metabolites for prediction of type 2 diabetes
.
J. Transl. Med.
19
:
393
.
25
Liu
,
H.
,
M.
Li
,
Y.
Song
,
W.
Xu
.
2018
.
TRIM21 restricts coxsackievirus B3 replication, cardiac and pancreatic injury via interacting with MAVS and positively regulating IRF3-mediated type-I interferon production
.
Front. Immunol.
9
:
2479
.
26
Chien
,
R. N.
,
Y. F.
Liaw
.
2022
.
Current trend in antiviral therapy for chronic hepatitis B
.
Viruses
14
:
434
.
27
Stoffel
,
W.
,
G.
Sticht
,
D.
LeKim
.
1968
.
Metabolism of sphingosine bases. IX. Degradation in vitro of dihydrospingosine and dihydrospingosine phosphate to palmitaldehyde and ethanolamine phosphate
.
Hoppe Seylers Z. Physiol. Chem.
349
:
1745
1748
.
28
Zhen
,
S.
,
L.
Hua
,
Y. H.
Liu
,
L. C.
Gao
,
J.
Fu
,
D. Y.
Wan
,
L. H.
Dong
,
H. F.
Song
,
X.
Gao
.
2015
.
Harnessing the clustered regularly interspaced short palindromic repeat (CRISPR)/CRISPR-associated Cas9 system to disrupt the hepatitis B virus
.
Gene Ther.
22
:
404
412
.
29
Lin
,
G.
,
K.
Zhang
,
J.
Li
.
2015
.
Application of CRISPR/Cas9 technology to HBV
.
Int. J. Mol. Sci.
16
:
26077
26086
.
30
Fauzyah
,
Y.
,
C.
Ono
,
S.
Torii
,
I.
Anzai
,
R.
Suzuki
,
T.
Izumi
,
Y.
Morioka
,
Y.
Maeda
,
T.
Okamoto
,
T.
Fukuhara
,
Y.
Matsuura
.
2021
.
Ponesimod suppresses hepatitis B virus infection by inhibiting endosome maturation
.
Antiviral Res.
186
:
104999
.
31
Grammatikos
,
G.
,
N.
Ferreiros
,
D.
Bon
,
S.
Schwalm
,
J.
Dietz
,
C.
Berkowski
,
D.
Fitting
,
E.
Herrmann
,
S.
Zeuzem
,
C.
Sarrazin
,
J.
Pfeilschifter
.
2015
.
Variations in serum sphingolipid levels associate with liver fibrosis progression and poor treatment outcome in hepatitis C virus but not hepatitis B virus infection
.
Hepatology
61
:
812
822
.
32
Rohrbach
,
T.
,
M.
Maceyka
,
S.
Spiegel
.
2017
.
Sphingosine kinase and sphingosine-1-phosphate in liver pathobiology
.
Crit. Rev. Biochem. Mol. Biol.
52
:
543
553
.
33
Mohammed
,
S.
,
A.
Bindu
,
A.
Viswanathan
,
K. B.
Harikumar
.
2023
.
Sphingosine 1-phosphate signaling during infection and immunity
.
Prog. Lipid Res.
92
:
101251
.
34
Chen
,
B.
,
X. P.
Chen
,
M. S.
Wu
,
W.
Cui
,
M.
Zhong
.
2014
.
Expressions of heparanase and upstream stimulatory factor in hepatocellular carcinoma
.
Eur. J. Med. Res.
19
:
45
.
35
Zeng
,
Y. L.
,
F.
Gao
,
C.
Zhang
,
P. P.
Ren
,
L.
Ma
,
X.
Wang
,
R.
Wang
,
Y.
Kang
,
K.
Li
.
2023
.
USF1 modulates transcription and cellular functions by regulating multiple transcription factors in Huh7 cells
.
Oncol. Lett.
26
:
532
.
36
Baidya
,
A.
,
M.
Khatun
,
R. K.
Mondal
,
S.
Ghosh
,
B. C.
Chakraborty
,
S.
Mallik
,
S. K. M.
Ahammed
,
A.
Chowdhury
,
S.
Banerjee
,
S.
Datta
.
2022
.
Hepatitis B virus suppresses complement C9 synthesis by limiting the availability of transcription factor USF-1 and inhibits formation of membrane attack complex: implications in disease pathogenesis
.
J. Biomed. Sci.
29
:
97
.
37
McGinley
,
M. P.
,
J. A.
Cohen
.
2021
.
Sphingosine 1-phosphate receptor modulators in multiple sclerosis and other conditions
.
Lancet
398
:
1184
1194
.
38
Motyl
,
J.
,
Ł.
Przykaza
,
P. M.
Boguszewski
,
P.
Kosson
,
J. B.
Strosznajder
.
2018
.
Pramipexole and fingolimod exert neuroprotection in a mouse model of Parkinson’s disease by activation of sphingosine kinase 1 and Akt kinase
.
Neuropharmacology
135
:
139
150
.
This article is distributed under The American Association of Immunologists, Inc., Reuse Terms and Conditions for Author Choice articles.

Supplementary data