Minireviews Open Access
Copyright ©The Author(s) 2016. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Sep 28, 2016; 22(36): 8161-8167
Published online Sep 28, 2016. doi: 10.3748/wjg.v22.i36.8161
Impact of hepatitis B virus infection on hepatic metabolic signaling pathway
Yi-Xian Shi, Chen-Jie Huang, Zheng-Gang Yang, State Key Lab of Diagnostic and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
Author contributions: Shi YX and Yang ZG specified the topic and wrote the paper; Huang CJ performed literature search and drafted the paper.
Supported by the National Natural Science Foundation of China, No. 81270500; The State 12th 5-Year Plan S&T Projects of China, No. 2012ZX10002007; and The National Basic Research Program (973 Program) in China, No. 2013CB531400.
Conflict-of-interest statement: The authors declared that there is no conflict of interest related to this study.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Zheng-Gang Yang, MD, PhD, State Key Lab of Diagnostic and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou 310003, Zhejiang Province, China. yangzg@zju.edu.cn
Telephone: +86-571-87236449 Fax: +86-571-87068731
Received: May 24, 2016
Peer-review started: May 25, 2016
First decision: July 13, 2016
Revised: August 1, 2016
Accepted: August 10, 2016
Article in press: August 10, 2016
Published online: September 28, 2016

Abstract

A growing body of epidemiologic research has demonstrated that metabolic derangement exists in patients with hepatitis B virus (HBV) infection, indicating that there are clinical associations between HBV infection and host metabolism. In order to understand the complex interplay between HBV and hepatic metabolism in greater depth, we systematically reviewed these alterations in different metabolic signaling pathways due to HBV infection. HBV infection interfered with most aspects of hepatic metabolic responses, including glucose, lipid, nucleic acid, bile acid and vitamin metabolism. Glucose and lipid metabolism is a particular focus due to the significant promotion of gluconeogenesis, glucose aerobic oxidation, the pentose phosphate pathway, fatty acid synthesis or oxidation, phospholipid and cholesterol biosynthesis affected by HBV. These altered metabolic pathways are involved in the pathological process of not only hepatitis B, but also metabolic disorders, increasing the occurrence of complications, such as hepatocellular carcinoma and liver steatosis. Thus, a clearer understanding of the hepatic metabolic pathways affected by HBV and its pathogenesis is necessary to develop more novel therapeutic strategies targeting viral eradication.

Key Words: Hepatitis B virus infection, Nucleic acid metabolism, Metabolic derangement, Metabolic signaling pathway, Glucose metabolism, Lipid metabolism, Bile acid metabolism, Vitamin metabolism

Core tip: Currently, hepatitis B virus (HBV) infection still poses a serious threat to public health, and causes approximately 1 million deaths annually due to HBV-related liver diseases. Thus, investigation into the complex host cellular responses to HBV infection is a crucial area of research. Multiple epidemiologic data have proved that patients with HBV infection often have metabolic disorders. Therefore, we systematically reviewed the alterations in metabolic response to HBV infection with regard to molecular mechanisms. Deciphering the detailed interplay mechanisms would contribute to our understanding of HBV-induced pathological processes and may lead to nutritional therapies as new anti-HBV treatments.



INTRODUCTION

Chronic hepatitis B (CHB) is a serious global health concern, which is estimated to affect approximately 350 million people worldwide and carries a significantly increased risk of serious liver disorders including liver cirrhosis, hepatic decompensation or hepatocellular carcinoma (HCC). Approximately, one million deaths occur each year due to CHB and its complications[1,2]. However, the intrinsic mechanisms of hepatitis B virus (HBV)-induced diseases are unclear, and no complete cure is currently available for CHB. Thus, an investigation of the complex host responses to HBV infection is a crucial area of research, which in turn could provide a more thorough understanding of the pathogenesis and potential novel targets in antiviral drug discovery.

The HBV genome (3.2 kb) is a partially double-stranded, relaxed circular DNA, mainly controlled through the transcriptional activation of four promoters (core, X, pre-S1 and pre-S2/S) and two enhancers (EnhI and EnhII)[3-6]. The recruitment of cellular transcription factors to the binding sites of HBV genome could regulate virus transcription. Some of these transcription factors are ubiquitous nuclear receptors such as C-AMP-response element binding protein, specificity protein 1, prospero-related homeobox protein and nuclear respiratory factor 1; some are liver-enriched nuclear receptors such as hepatocyte nuclear factor 4, alpha (HNF4a), CAAT enhancer-binding protein, peroxisome proliferator-activated receptors, alpha/retinoid X receptors, alpha (PPARa/RXRa) and farnesoid X receptor (FXR)[7,8].

Interestingly, the native role of most HBV-bound transcription factors is the coordination and control of hepatic metabolism[8]. For example, HNF4a plays a key role in glucose metabolism in the liver[9]. PPARa controls fatty acid β-oxidation and is a crucial regulator of genes involved in the cellular fasting response[10]. FXR, activated by bile acids, is a molecular link between lipid metabolism and bile acid[11]. Thus, it indicates that HBV has adopted a smart mode of regulation, which is similar to that of major hepatic metabolic genes, implying that there is an association between metabolism and HBV infection. Our previous work also suggested that activation of fatty acid oxidation-associated PPARα was required for fasting-induced HBV transcription[12].

Accumulating epidemiologic evidence has shown that there is still debate regarding the clinical associations between HBV infection and host metabolism. For instance, patients with chronic HBV infection, compared with healthy adults, have lower triglyceride (TG) and high-density lipoprotein (HDL) levels, but a higher adiponectin level[13]. A review by Janicko et al[14] described strong correlations between CHB and the metabolic syndrome, non-alcoholic fatty liver disease or dyslipidemia, whereas an inconclusive association between diabetes mellitus and CHB has also been described. However, in metabolic signaling pathways, an increasing number of studies have shown that HBV modulates all aspects of host hepatic metabolism. In order to understand the unique interplay between HBV and hepatic metabolism in greater depth, we systematically reviewed these alterations in metabolic signaling pathways due to HBV infection.

HBV AND GLUCOSE METABOLISM

Glucose homeostasis is regulated by balancing the output and the storage of glucose[15]. Glucose metabolism in hepatocytes can be divided broadly into two categories: anabolism and catabolism, including gluconeogenesis, glycolysis, aerobic oxidation and the pentose phosphate pathway.

Previous studies indicated that HBV infection could affect either gluconeogenesis or glucose aerobic oxidation. According to the study by Park[16], hepatitis B virus X protein (HBx) functions as an important positive regulator of gluconeogenesis. In HBx-overexpressing (HBxTg) mice and inducible nitric oxide synthase (iNOS)-knocked out HBxTg mice, increased HBx expression significantly up-regulated the gene expression of hepatic key gluconeogenic enzymes (PEPCK, G6Pase) and the production of hepatic glucose, leading to hyperglycemia and impaired glucose tolerance. These effects are considered to be mediated through the nitric oxide (NO)/JNK pathway. However, other studies have demonstrated that HBV can promote glucose aerobic oxidation. By combining proteomics, metabolomics and molecular biological assays in HepG2.2.15 and HepG2 cell models, Li et al[17] provided a holistic view of the interplay between host metabolism and HBV. They pointed out that enzymes which regulate the glycolysis pathway, such as fructose-bisphosphate aldolase, alpha enolase, triosephosphate isomerase, phosphoglycerate kinase 1 and glucose-6-phosphate isomerase, and enzymes involved in the tricarboxylic acid (TCA) cycle, including malate dehydrogenase, citrate synthase and succinate dehydrogenase, are all significantly up-regulated in HepG2.2.15 cells, subsequently leading to elevated levels of corresponding intermediates, such as lactate in glycolysis and fumarate, succinate and 2-oxoglutarate in the TCA cycle. These data suggested that glycolysis and the TCA cycle are stimulated in host cells due to HBV infection. In addition, another study revealed that a HBV pre-S2 mutant could induce aerobic oxidation via activation of MTOR signaling, which may contribute to HBV tumorigenesis[18].

Furthermore, HBV infection could promote the pentose phosphate pathway (PPP). Overexpression of HBx caused the nuclear translocation and activation of NF-E2-related factor 2, resulting in up-regulation of glucose-6-phosphate dehydrogenase, which is the first and rate-limiting enzyme of the PPP converting glucose-6-phosphate into 6-phosphogluconolactone[19]. Enhancement of the PPP by HBx-mediated elevation of G6PD provided host cells with more ribose for nucleotide biosynthesis to support their proliferation, which might contribute to HBV-associated hepatocarcinogenesis. The change in G6PD was also supported by a systems biology model[17]. It was reported that G6PD participating in the PPP was markedly increased, accompanied by elevated nucleotide levels, such as AMP, ADP, uridine 59-diphosphate and inosine-59-monophosphate.

HBV AND LIPID METABOLISM

The liver, the main organ for the synthesis and circulation of lipids (e.g., fatty acids, fats, phospholipids and cholesterol), oxidation of fatty acids and the production of ketone bodies, plays an important role in lipid metabolism[20].

A significant amount of basic research has indicated that HBV infection has an effect on fatty acid metabolism. Many, but not all, studies have shown that HBV can promote the synthesis of fatty acids. Based on HPLC/MS analysis and two-dimensional electrophoresis (2-DE), fatty acid binding 5 and Acyl-CoA binding protein implicated in fatty acid metabolism and synthesis, which can bind Acyl-CoA and fatty acids with high affinity, are markedly increased in hepatitis B virus transgenic mice (HBV-Tg mice)[21]. HBV-influenced genes in lipid biosynthetic pathways in HBV-Tg mice were identified by cDNA microarray analysis, in which retinol binding protein 1 (RBP1), sterol regulatory element binding protein 2 (SREBP2), ATP citrate lyase and fatty acid synthase (FAS) were all strongly upregulated[22]. However, in contrast to the above studies, Wang et al[23] recently proposed that up-regulation of HBx could facilitate fatty acid oxidation (FAO) and subsequently maintain intracellular NADPH and ATP levels under glucose deprivation, which is of great importance for HCC cell survival under conditions of metabolic stress.

Recently, accumulating evidence from experimental investigations has suggested that HBV infection is a potential trigger of liver steatosis. HBx can induce hepatic steatosis at all aspects, such as increasing fatty acid binding, promoting lipid synthesis and inhibiting secretion of apolipoprotein (Figure 1). Fatty acid binding protein 1 (FABP1), responsible for the uptake, metabolism and transport of long-chain fatty acids (LFA)[24], plays a key role in intracellular fatty acid utilization and transport[25]. Forced expression of HBx induced liver steatosis through up-regulation of FABP1, whereas gene silencing of FABP1 blocked lipid accumulation in both in vivo and in vitro models[26]. LXR, SREBP1 and PPARγ are master regulators in hepatic lipogenesis: LXR directly induces expression of SREBP1, which up-regulates lipogenic genes[27]; activation of LXR also stimulates adipocyte differentiation through induction of PPARγ expression[28]. Both are suggested to be of vital importance in hepatic lipid accumulation. Several studies have demonstrated that HBx increased the gene expression and transcriptional activity of LXR-mediated SREBP1 and PPARγ, thereby inducing the expression of hepatic lipogenic genes (fatty acid synthase, stearoyl-CoA desaturase, acetyl-CoA carboxylase) and adipogenic genes (adipsin, adiponectin, aP2/adipose fatty acid–binding protein), finally accompanied by the accumulation of lipid droplets[29-32]. Apolipoprotein B (apoB), required for the secretion and assembly of low-density lipoproteins (LDL) and very low-density lipoproteins (VLDL), is assembled into a secretion-competent particle with lipids[33-35]. It has been reported that HBx mediated aberrantly glycosylated apoB by elevating the expression of N-Acetylglucosaminyltransferase III (GnT-III) resulted in inhibition of apoB secretion as well as intracellular accumulation of cholesterol and triglyceride[36].

Figure 1
Figure 1 The molecular mechanisms contributing to liver steatosis following hepatitis B virus infection. Hepatitis B virus (HBV) infection can induce the accumulation of lipids via three different regulatory mechanisms, including elevated expression of FABP1, up-regulation of LXR, SREBP1 and PPARγ and increased expression of GnT-III. On the one hand, up-regulation of FABP1 would increase fatty acid binding and transport. On the other hand, induction of LXR-mediated SREBP1 and PPARγ would result in increased transcriptional activity of hepatic lipogenic genes (FAS, SCD, ACC) and adipogenic genes (adipoQ, adipsin, AP2). In addition, elevation of GnT-III would cause glycosylation and dysfunction of apoB, finally leading to reduced secretion of VLDL (containing apoB, CHO and TG). LFA: Long-chain fatty acids; FABP1: Fatty acid binding protein 1; FAS: Fatty acid synthase; SCD: Stearoyl-CoA desaturase; ACC: Acetyl-CoA carboxylase; adipoQ: Adiponectin; AP2: aP2/adipose fatty acid–binding protein; apoB: Apolipoprotein B; GnT-III: N-Acetylglucosaminyltransferase III; CHO: Cholesterol; TG: Triglyceride; VLDL: Very low density lipoproteins.

In addition, phospholipid and cholesterol metabolism are also altered by the presence of HBV. Phosphatidylcholine (PC) is a major component of the biological membrane[37], and acts as a precursor for the synthesis of lipid signaling molecules[38]. In comparison with HepG2, the key enzymes participating in PC synthesis, such as choline-phosphate cytidylyltransferase A, choline kinase a, choline phosphotransferase 1 and choline/ethanolamine phosphotransferase 1 are all up-regulated in HepG2.2.15 cells, consistent with the elevated levels of phosphocholine and reduced levels of choline[17]. These results strongly indicate that HBV infection can promote the biosynthesis of PC. Cholesterol, a type of lipid different from triglyceride and phospholipid, has two essential metabolic fates: conversion into bile acids or steroid hormones[39]. HBV-infected humanized mice displayed a significant increase in human genes related to the uptake, biosynthesis, and transcriptional regulation of cholesterol, such as low-density lipoprotein receptor (LDLR), hydroxymethylglutaryl-coenzyme A reductase (HMGCR) and SREBP2[40]. Another study provided evidence that HBV exacerbated hepatic cholesterol accumulation via up-regulation of LDLR and HMGCR in HepG2 cells[41].

HBV AND NUCLEIC ACID METABOLISM

It is well known that the main function of the nucleotide is the biosynthesis of nucleic acids. Many studies have reported that DNA damage can cause abnormalities in nucleic acid metabolism[42]. HBV infection also influences this process via HBx-induced DNA damage, which may result in the onset of hepatocarcinogenesis[43]. Thus, identifying the distinguishing nucleic acid metabolites under HBx induction may help to understand the occurrence of HCC. A new study[44] using a systematic approach combining metabonomics and mRNA microarray analysis indicated that HBx could initially induce DNA damage and then disrupt nucleic acid metabolism, resulting in a significant decrease in DNA damage-related genes (BRCA1, TP53, RPA1, DDB1, TCEA1 and NHEJ1) and intermediates of nucleic acid metabolism (guanosine, inosine and uridine), which in turn blocked DNA repair and probably contributed to the development of HCC.

HBV AND BILE ACID METABOLISM

Bile acid, mainly synthesized in the liver from cholesterol, plays a key role in the digestion and absorption of lipids[45]. Human NTCP (SLC10A1), located in the basolateral membrane of hepatocytes, functions as a main transporter to mediate entry of bile salts from portal blood into hepatocytes[46].

Recently, the detection of NTCP, acting as a functional entry receptor for HBV, clearly represents a typical milestone in our knowledge of HBV infection[47]. HBV exploits NTCP for species-specific entry into hepatocytes[48]. Hence, emerging evidence demonstrated the probable association between HBV infection and bile acid. Yan et al[49] showed that the HBV pre-S1 lipopeptide efficiently blocked the uptake of bile salts by NTCP, suggesting that HBV infection may limit the physiological function of NTCP. Reduced bile salts could promote compensatory bile acid synthesis to maintain its homeostasis[50,51]. This compensation was confirmed by the strong induction of hCYP7A1 (the rate-limiting enzyme converting cholesterol to bile acid), decreased FXR (the positive transcription factor of SHP) nuclear translocation and significant reduction of SHP (the corepressor of hCYP7A1 transcription) in human liver-chimeric uPA/SCID mice infected with HBV[40].

HBV AND VITAMIN METABOLISM

Vitamin A, including retinol, retinal and retinoic acid, plays a critical role in visual function as well as cell growth and differentiation[52]. Previous data provided evidence that retinoic acid could enhance HBV transcription and replication through activation of RXRa[53,54]. Most interestingly, another study demonstrated that HBV infection could promote retinol metabolism-related proteins RBP, CRBP1 and ALDH1 as shown by 2-DE and MS/MS analysis[55]. It is reasonable that more retinol would be pumped into cells and converted into retinoic acid during HBV infection. HBV infection may up-regulate retinoic acid by promoting retinol metabolism and thereby facilitating self- replication through activation of RXRa, leading to an increased risk of liver damage, which was considered a positive feedback[56].

Vitamin D, including its bioactive vitamin D metabolite [1,25(OH)2D3] and stable, easy-to-quantify metabolite (25(OH)D3), plays an emerging role in metabolic and inflammatory liver diseases[57]. A study has demonstrated a significant association between low levels of serum 25(OH)D3 and high HBV DNA levels in CHB patients[57]. However, the molecular mechanism underlying inverse seasonal fluctuations of HBV DNA and 25(OH)D3 serum levels still remains to be elucidated.

CONCLUSION

In conclusion, we have systematically outlined the hepatic metabolic responses to HBV infection in this review. According to the above observations, multiple studies combining systematic approaches and molecular biological assays found that, from the molecular mechanism perspective, HBV infection interfered with the hepatic metabolic signaling pathway (Figure 2), including glucose, lipid, nucleic acid, bile acid and vitamin metabolism, ultimately resulting in metabolic derangement. Furthermore, these altered metabolic pathways may also contribute to the pathological processes of other HBV-induced diseases, such as hepatocellular carcinoma. Therefore, in this review, deciphering the molecular mechanisms of the metabolic pathways during HBV infection has shed new light on the pathological processes, and provides a new, revolutionary, potential means of directly fighting against this virus.

Figure 2
Figure 2 Changes in the hepatic metabolic signaling pathway induced by hepatitis B virus infection. Alterations in related signaling pathways (including glucose, lipids, nucleic acids, bile acids and vitamins) following hepatitis B virus (HBV) infection are marked and highlighted in this figure. The influence of HBV infection on vitamin D metabolism is unclear.
Footnotes

Manuscript source: Invited manuscript

Specialty type: Gastroenterology and hepatology

Country of origin: China

Peer-review report classification

Grade A (Excellent): A, A

Grade B (Very good): 0

Grade C (Good): 0

Grade D (Fair): 0

Grade E (Poor): 0

P- Reviewer: Ebert G, Ohkoshi S S- Editor: Yu J L- Editor: A E- Editor: Wang CH

References
1.  Dandri M, Locarnini S. New insight in the pathobiology of hepatitis B virus infection. Gut. 2012;61 Suppl 1:i6-17.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
2.  Ganem D, Prince AM. Hepatitis B virus infection--natural history and clinical consequences. N Engl J Med. 2004;350:1118-1129.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Pang PK, Wang R, Shan J, Karpinski E, Benishin CG. Specific inhibition of long-lasting, L-type calcium channels by synthetic parathyroid hormone. Proc Natl Acad Sci USA. 1990;87:623-627.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Shaul Y, Rutter WJ, Laub O. A human hepatitis B viral enhancer element. EMBO J. 1985;4:427-430.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Wang Y, Chen P, Wu X, Sun AL, Wang H, Zhu YA, Li ZP. A new enhancer element, ENII, identified in the X gene of hepatitis B virus. J Virol. 1990;64:3977-3981.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Yee JK. A liver-specific enhancer in the core promoter region of human hepatitis B virus. Science. 1989;246:658-661.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Quasdorff M, Protzer U. Control of hepatitis B virus at the level of transcription. J Viral Hepat. 2010;17:527-536.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 150]  [Cited by in F6Publishing: 1]  [Article Influence: 0.1]  [Reference Citation Analysis (0)]
8.  Bar-Yishay I, Shaul Y, Shlomai A. Hepatocyte metabolic signalling pathways and regulation of hepatitis B virus expression. Liver Int. 2011;31:282-290.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 3]  [Reference Citation Analysis (0)]
9.  Rhee J, Inoue Y, Yoon JC, Puigserver P, Fan M, Gonzalez FJ, Spiegelman BM. Regulation of hepatic fasting response by PPARgamma coactivator-1alpha (PGC-1): requirement for hepatocyte nuclear factor 4alpha in gluconeogenesis. Proc Natl Acad Sci USA. 2003;100:4012-4017.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Leone TC, Weinheimer CJ, Kelly DP. A critical role for the peroxisome proliferator-activated receptor alpha (PPARalpha) in the cellular fasting response: the PPARalpha-null mouse as a model of fatty acid oxidation disorders. Proc Natl Acad Sci USA. 1999;96:7473-7478.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Claudel T, Staels B, Kuipers F. The Farnesoid X receptor: a molecular link between bile acid and lipid and glucose metabolism. Arterioscler Thromb Vasc Biol. 2005;25:2020-2030.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Shi Y, Li Y, Huang C, Ying L, Xue J, Wu H, Chen Z, Yang Z. Resveratrol enhances HBV replication through activating Sirt1-PGC-1α-PPARα pathway. Sci Rep. 2016;6:24744.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 33]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
13.  Hsu CS, Liu CH, Wang CC, Tseng TC, Liu CJ, Chen CL, Chen PJ, Chen DS, Kao JH. Impact of hepatitis B virus infection on metabolic profiles and modifying factors. J Viral Hepat. 2012;19:e48-e57.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 48]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
14.  Jarcuska P, Drazilova S, Fedacko J, Pella D, Janicko M. Association between hepatitis B and metabolic syndrome: Current state of the art. World J Gastroenterol. 2016;22:155-164.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
15.  Klover PJ, Mooney RA. Hepatocytes: critical for glucose homeostasis. Int J Biochem Cell Biol. 2004;36:753-758.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Shin HJ, Park YH, Kim SU, Moon HB, Park DS, Han YH, Lee CH, Lee DS, Song IS, Lee DH. Hepatitis B virus X protein regulates hepatic glucose homeostasis via activation of inducible nitric oxide synthase. J Biol Chem. 2011;286:29872-29881.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 41]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
17.  Li H, Zhu W, Zhang L, Lei H, Wu X, Guo L, Chen X, Wang Y, Tang H. The metabolic responses to hepatitis B virus infection shed new light on pathogenesis and targets for treatment. Sci Rep. 2015;5:8421.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 92]  [Article Influence: 10.2]  [Reference Citation Analysis (0)]
18.  Teng CF, Hsieh WC, Wu HC, Lin YJ, Tsai HW, Huang W, Su IJ. Hepatitis B Virus Pre-S2 Mutant Induces Aerobic Glycolysis through Mammalian Target of Rapamycin Signal Cascade. PLoS One. 2015;10:e0122373.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 36]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
19.  Liu B, Fang M, He Z, Cui D, Jia S, Lin X, Xu X, Zhou T, Liu W. Hepatitis B virus stimulates G6PD expression through HBx-mediated Nrf2 activation. Cell Death Dis. 2015;6:e1980.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 70]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
20.  Nguyen P, Leray V, Diez M, Serisier S, Le Bloc’h J, Siliart B, Dumon H. Liver lipid metabolism. J Anim Physiol Anim Nutr (Berl). 2008;92:272-283.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 471]  [Cited by in F6Publishing: 551]  [Article Influence: 34.4]  [Reference Citation Analysis (1)]
21.  Yang F, Yan S, He Y, Wang F, Song S, Guo Y, Zhou Q, Wang Y, Lin Z, Yang Y. Expression of hepatitis B virus proteins in transgenic mice alters lipid metabolism and induces oxidative stress in the liver. J Hepatol. 2008;48:12-19.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 68]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
22.  Hajjou M, Norel R, Carver R, Marion P, Cullen J, Rogler LE, Rogler CE. cDNA microarray analysis of HBV transgenic mouse liver identifies genes in lipid biosynthetic and growth control pathways affected by HBV. J Med Virol. 2005;77:57-65.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 58]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
23.  Wang MD, Wu H, Huang S, Zhang HL, Qin CJ, Zhao LH, Fu GB, Zhou X, Wang XM, Tang L. HBx regulates fatty acid oxidation to promote hepatocellular carcinoma survival during metabolic stress. Oncotarget. 2016;7:6711-6726.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Hertzel AV, Bernlohr DA. The mammalian fatty acid-binding protein multigene family: molecular and genetic insights into function. Trends Endocrinol Metab. 2000;11:175-180.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Schroeder F, Jolly CA, Cho TH, Frolov A. Fatty acid binding protein isoforms: structure and function. Chem Phys Lipids. 1998;92:1-25.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Wu YL, Peng XE, Zhu YB, Yan XL, Chen WN, Lin X. Hepatitis B Virus X Protein Induces Hepatic Steatosis by Enhancing the Expression of Liver Fatty Acid Binding Protein. J Virol. 2016;90:1729-1740.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 71]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
27.  Chen G, Liang G, Ou J, Goldstein JL, Brown MS. Central role for liver X receptor in insulin-mediated activation of Srebp-1c transcription and stimulation of fatty acid synthesis in liver. Proc Natl Acad Sci USA. 2004;101:11245-11250.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Seo JB, Moon HM, Kim WS, Lee YS, Jeong HW, Yoo EJ, Ham J, Kang H, Park MG, Steffensen KR. Activated liver X receptors stimulate adipocyte differentiation through induction of peroxisome proliferator-activated receptor gamma expression. Mol Cell Biol. 2004;24:3430-3444.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Kim KH, Shin HJ, Kim K, Choi HM, Rhee SH, Moon HB, Kim HH, Yang US, Yu DY, Cheong J. Hepatitis B virus X protein induces hepatic steatosis via transcriptional activation of SREBP1 and PPARgamma. Gastroenterology. 2007;132:1955-1967.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 209]  [Cited by in F6Publishing: 210]  [Article Influence: 12.4]  [Reference Citation Analysis (0)]
30.  Shieh YS, Chang YS, Hong JR, Chen LJ, Jou LK, Hsu CC, Her GM. Increase of hepatic fat accumulation by liver specific expression of Hepatitis B virus X protein in zebrafish. Biochim Biophys Acta. 2010;1801:721-730.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 48]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
31.  Kim K, Kim KH, Kim HH, Cheong J. Hepatitis B virus X protein induces lipogenic transcription factor SREBP1 and fatty acid synthase through the activation of nuclear receptor LXRalpha. Biochem J. 2008;416:219-230.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 81]  [Cited by in F6Publishing: 91]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
32.  Na TY, Shin YK, Roh KJ, Kang SA, Hong I, Oh SJ, Seong JK, Park CK, Choi YL, Lee MO. Liver X receptor mediates hepatitis B virus X protein-induced lipogenesis in hepatitis B virus-associated hepatocellular carcinoma. Hepatology. 2009;49:1122-1131.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 118]  [Cited by in F6Publishing: 124]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
33.  Yao Z, McLeod RS. Synthesis and secretion of hepatic apolipoprotein B-containing lipoproteins. Biochim Biophys Acta. 1994;1212:152-166.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Ginsberg HN. Role of lipid synthesis, chaperone proteins and proteasomes in the assembly and secretion of apoprotein B-containing lipoproteins from cultured liver cells. Clin Exp Pharmacol Physiol. 1997;24:A29-A32.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Fisher EA, Ginsberg HN. Complexity in the secretory pathway: the assembly and secretion of apolipoprotein B-containing lipoproteins. J Biol Chem. 2002;277:17377-17380.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Kang SK, Chung TW, Lee JY, Lee YC, Morton RE, Kim CH. The hepatitis B virus X protein inhibits secretion of apolipoprotein B by enhancing the expression of N-acetylglucosaminyltransferase III. J Biol Chem. 2004;279:28106-28112.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 59]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
37.  Esko JD, Nishijima M, Raetz CR. Animal cells dependent on exogenous phosphatidylcholine for membrane biogenesis. Proc Natl Acad Sci USA. 1982;79:1698-1702.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Yalcin A, Clem B, Makoni S, Clem A, Nelson K, Thornburg J, Siow D, Lane AN, Brock SE, Goswami U. Selective inhibition of choline kinase simultaneously attenuates MAPK and PI3K/AKT signaling. Oncogene. 2010;29:139-149.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
39.  Peet DJ, Turley SD, Ma W, Janowski BA, Lobaccaro JM, Hammer RE, Mangelsdorf DJ. Cholesterol and bile acid metabolism are impaired in mice lacking the nuclear oxysterol receptor LXR alpha. Cell. 1998;93:693-704.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Oehler N, Volz T, Bhadra OD, Kah J, Allweiss L, Giersch K, Bierwolf J, Riecken K, Pollok JM, Lohse AW. Binding of hepatitis B virus to its cellular receptor alters the expression profile of genes of bile acid metabolism. Hepatology. 2014;60:1483-1493.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 105]  [Cited by in F6Publishing: 105]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
41.  Li YJ, Zhu P, Liang Y, Yin WG, Xiao JH. Hepatitis B virus induces expression of cholesterol metabolism-related genes via TLR2 in HepG2 cells. World J Gastroenterol. 2013;19:2262-2269.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 28]  [Cited by in F6Publishing: 27]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
42.  Zheng G, Fu Y, He C. Nucleic acid oxidation in DNA damage repair and epigenetics. Chem Rev. 2014;114:4602-4620.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
43.  Na TY, Ka NL, Rhee H, Kyeong D, Kim MH, Seong JK, Park YN, Lee MO. Interaction of hepatitis B virus X protein with PARP1 results in inhibition of DNA repair in hepatocellular carcinoma. Oncogene. 2016; Epub ahead of print.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Dan Yue Y, Cheng L, Ma J, Xi Y, Yang L, Su C, Shao B, Huang A, Xiang R, Cheng P. Hepatitis B virus X protein (HBx)-induced abnormalities of nucleic acid metabolism revealed by (1)H-NMR-based metabonomics. Sci Rep. 2016;6:24430.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 34]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
45.  Chiang JY. Bile acids: regulation of synthesis. J Lipid Res. 2009;50:1955-1966.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1034]  [Cited by in F6Publishing: 1112]  [Article Influence: 74.1]  [Reference Citation Analysis (0)]
46.  Stieger B. The role of the sodium-taurocholate cotransporting polypeptide (NTCP) and of the bile salt export pump (BSEP) in physiology and pathophysiology of bile formation. Handb Exp Pharmacol. 2011;205-259.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
47.  Yan H, Zhong G, Xu G, He W, Jing Z, Gao Z, Huang Y, Qi Y, Peng B, Wang H. Sodium taurocholate cotransporting polypeptide is a functional receptor for human hepatitis B and D virus. Elife. 2012;1:e00049.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1280]  [Cited by in F6Publishing: 1422]  [Article Influence: 118.5]  [Reference Citation Analysis (0)]
48.  Ni Y, Lempp FA, Mehrle S, Nkongolo S, Kaufman C, Fälth M, Stindt J, Königer C, Nassal M, Kubitz R. Hepatitis B and D viruses exploit sodium taurocholate co-transporting polypeptide for species-specific entry into hepatocytes. Gastroenterology. 2014;146:1070-1083.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 540]  [Cited by in F6Publishing: 548]  [Article Influence: 54.8]  [Reference Citation Analysis (0)]
49.  Yan H, Peng B, Liu Y, Xu G, He W, Ren B, Jing Z, Sui J, Li W. Viral entry of hepatitis B and D viruses and bile salts transportation share common molecular determinants on sodium taurocholate cotransporting polypeptide. J Virol. 2014;88:3273-3284.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 178]  [Cited by in F6Publishing: 178]  [Article Influence: 17.8]  [Reference Citation Analysis (0)]
50.  Patman G. Hepatitis: HBV infection alters bile acid metabolism gene profile. Nat Rev Gastroenterol Hepatol. 2014;11:332.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 3]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
51.  Geier A. Hepatitis B virus: the “metabolovirus” highjacks cholesterol and bile acid metabolism. Hepatology. 2014;60:1458-1460.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 27]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
52.  Dowling JE, Wald G. THE BIOLOGICAL FUNCTION OF VITAMIN A ACID. Proc Natl Acad Sci USA. 1960;46:587-608.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Huan B, Kosovsky MJ, Siddiqui A. Retinoid X receptor alpha transactivates the hepatitis B virus enhancer 1 element by forming a heterodimeric complex with the peroxisome proliferator-activated receptor. J Virol. 1995;69:547-551.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Huan B, Siddiqui A. Retinoid X receptor RXR alpha binds to and trans-activates the hepatitis B virus enhancer. Proc Natl Acad Sci USA. 1992;89:9059-9063.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Tong A, Wu L, Lin Q, Lau QC, Zhao X, Li J, Chen P, Chen L, Tang H, Huang C. Proteomic analysis of cellular protein alterations using a hepatitis B virus-producing cellular model. Proteomics. 2008;8:2012-2023.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 63]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
56.  Mawson AR, Steele TA. Possible role of retinoids in hepatitis B virus-associated liver damage. Exp Biol Med (Maywood). 2001;226:734-739.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Farnik H, Bojunga J, Berger A, Allwinn R, Waidmann O, Kronenberger B, Keppler OT, Zeuzem S, Sarrazin C, Lange CM. Low vitamin D serum concentration is associated with high levels of hepatitis B virus replication in chronically infected patients. Hepatology. 2013;58:1270-1276.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 96]  [Cited by in F6Publishing: 104]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]