The cellular mechanisms by which hepatitis B virus (HBV) is assembled and exported are largely undefined. Recently, it has been suggested that these steps require the multivesicular body (MVB) and the autophagic machinery. However, the mechanisms by which HBV might regulate these compartments are unclear. In this study, we have found that by activating Rab7a, HBV alters its own secretion by inducing dramatic changes in the morphology of MVB and autophagic compartments. These changes are characterized by the formation of numerous tubules that are dependent upon the increase in Rab7 activity observed in the HBV‐expressing HepG2.2.15 cells compared to HepG2 cells. Interestingly, transfection‐based expression of the five individual viral proteins indicated that the precore protein, which is a precursor of HBeAg, was largely responsible for the increased Rab7 activity. Finally, small interfering RNA (siRNA)‐mediated depletion of Rab7 significantly increased the secretion of virions, suggesting that reduced delivery of the virus to the lysosome facilitates viral secretion. These findings provide novel evidence indicating that HBV can regulate its own secretion through an activation of the endo‐lysosomal and autophagic pathway mediated by Rab7 activation.

Hepatitis B virus (HBV) infection is a global health problem as infected individuals are at risk of chronic hepatitis, liver cirrhosis and hepatocellular carcinoma. It is estimated that 350–400 million people are chronically infected with HBV (McMahon, 2005), despite the availability of effective vaccines. Although nucleoside and nucleotide analogues and peginterferon are used to prevent the progression as standard therapies (Rijckborst et al., 2011), these drugs cannot eradicate HBV completely and their efficacies are limited. Thus, a better understanding of the HBV life cycle and the mechanisms by which it usurps established host cell pathways is essential to enable the identification of new antiviral targets.

The infectious HBV virion (or Dane particle) is a spherical particle that is 42 nm in diameter. The virion consists of an inner icosahedral nucleocapsid, which contains circular partially double‐stranded genomic DNA, and an outer envelope composed of cellular lipids and three HBV surface proteins [small (SHBs), medium (MHBs) and large (LHBs)]. These surface proteins form also an excess of empty subviral particles (SVPs). Although the mature nucleocapsid is known to originate in the cytosol and contains the HBV core protein (HBc) (Bruss, 2007), the mechanisms by which the nucleocapsid is assembled, enveloped within the endoplasmic reticulum (ER) membrane (Eble et al., 1987) and trafficked to the cell surface for release are poorly understood. Recently, a compartment of the late endocytic pathway, the multivesicular body (MVB), has been shown to participate in the final stages of HBV maturation and release. The HBV appears to reside transiently in the MVB and disruption of this compartment results in a decrease of released HBV (Kian Chua et al., 2006; Lambert et al., 2007; Stieler and Prange, 2014; Watanabe et al., 2007). Although the structural interactions between the HBV and this organelle remain unclear, it appears that virus buds inwards into invaginations of the MVB limiting membrane, forming intraluminal vesicles (ILVs), a process that is topologically equivalent to enveloped viral budding from the cell surface (Prange, 2012). Subsequently, the MVBs are believed to dock at the hepatocyte surface and release their intraluminal viral cargo in a process that might resemble exosomal release (Hanson and Cashikar, 2012). More recently, the autophagic machinery has been implicated in HBV replication both morphologically and functionally, as targeted knockdowns of the autophagic proteins Atg5, Atg7 or Beclin1 reduce HBV release (Li et al., 2011; Sir et al., 2010; Tian et al., 2011). A physical and dynamic relationship is known to exist between MVBs and autophagosomes as they appear to interact (Berg et al., 1998; Fader et al., 2008) prior to fusion with a terminal lysosome and exchange a variety of different components (Fader and Colombo, 2009). The mechanisms that support the interactions between these organelles are a topic of intense study, and the processes by which the HBV utilizes these organelles to its own end are almost completely undefined.

The Rab proteins are small GTPases that act as molecular switches, orchestrating the formation, transport, tethering and fusion of vesicles in the secretory and endocytic pathways. As for most switch enzymes, the GTP‐bound state of a Rab regulates its binding to a variety of distinct effector proteins that facilitate vesicular traffic (Stenmark, 2009). In humans, there are more than 60 Rabs, many with unknown functions, although Rab7 (for which there are two isoforms, Rab7a and Rab7b) has been demonstrated to play a central role in regulating endo‐lysosomal membrane traffic (Bucci et al., 2000; Cantalupo et al., 2001). Rab7 is required for the maturation of late endosomes/MVBs and also autophagosomes, directing the trafficking of cargos along microtubules and participating in the fusion step with lysosomes (Hyttinen et al., 2013; Vanlandingham and Ceresa, 2009). We hypothesized that Rab7 could play an important role in the regulation of HBV assembly through the late endocytic pathway of hepatocytes. In this study, we show that the HBV itself is capable of activating Rab7 leading to the formation of pronounced tubular networks extending from MVBs and autophagosomes. These tubules appear to promote the fusion of MVBs, autophagosomes and lysosomes and facilitate the degradation of HBV. These findings indicate that the HBV alters the endo‐lysosomal and autophagic pathways through the regulation of a specific Rab protein and provides novel insights into host–pathogen interactions.

HBV resides within markedly tubulated Rab7‐associated MVB compartments

Based on the findings of others, HBV appears to utilize the MVB (Kian Chua et al., 2006; Lambert et al., 2007; Watanabe et al., 2007) and autophagic compartments (Li et al., 2011; Sir et al., 2010; Tian et al., 2011) during assembly, maturation and release. As Rab7 is known to regulate the dynamics and traffic between these late endocytic compartments and the lysosome (Cantalupo et al., 2001; Jäger et al., 2004; Stenmark, 2009), we tested whether alterations in Rab7 expression and/or function might interfere with the viral life cycle. First, HepG2.2.15 cells, which stably express HBV, but are not re‐infectable with released virus, were transfected with GFP‐tagged wild‐type Rab7 (GFP–Rab7wt) and the relationship between the virus and the Rab7‐positive compartment was assessed by confocal microscopy. Rab7 localized along numerous large punctate late endosomes and MVBs that also contained large quantities of HBV proteins (LHBs and HBc; Fig. 1A,B). As expected, mCherry‐tagged Rab‐interacting lysosomal protein (RILP), which is a classic effector protein of active Rab7 and has been used to visualize active Rab7 in the cell (Cantalupo et al., 2001), also colocalized with these HBV proteins (supplementary material Fig. S1A). We further confirmed that HBV proteins reside within MVBs and other late endosomal structures by colocalization studies of these viral antigens in compartments positive for the GFP‐tagged autophagosome marker LC3 (also known as MAP1LC3) and GFP‐tagged LAMP1 as a lysosomal marker (Fig. 1A,B). In addition, cells were stained with antibodies against the MVB marker Hrs, the tetraspanin MVB marker CD63 or the autophagosomal protein LC3. Cells were then co‐stained with for the viral LHBs (supplementary material Fig. S1B–D). In addition, transmission electron microscopy (TEM) images of HepG2.2.15 showed numerous electron‐dense virus‐like particles of 40 nm in diameter residing within both MVBs and autophagosome compartments (Fig. 1C,D). However, these virus‐like structures were not observed in the untransfected HepG2 cells (data not shown). These particles do not look like the typical Dane particles, which are observed with negative staining. It was considered that this is due to the difference in staining technique.

Fig. 1.

HBV components colocalize with MVBs and autophagosomes of virus‐expressing hepatocytes. (A,B) HepG2.2.15 cells were transfected to express different GFP‐tagged late endosome, MVB, lysosome or autophagosome markers including Rab7 (GFP–Rab7wt), GFP–LC3 and GFP–LAMP1 (green). Cells were double labeled with antibodies to two HBV antigens including anti‐PreS1 of LHBs (A) or an anti‐HBc antibody (red) (B), then analyzed by confocal microscopy. The viral antigens can be found in small putative MVB compartments (Rab7‐positive) but appear most prominent in large autophagosome organelles positive for LAMP1 and LC3. Scale bars: 10 µm. (C,D) Transmission electron microscopy (TEM) of HepG2.2.15 cells showing electron‐dense virus‐like particles (arrows) in MVBs (C) and in a large autophagosome (AP) (D).

Fig. 1.

HBV components colocalize with MVBs and autophagosomes of virus‐expressing hepatocytes. (A,B) HepG2.2.15 cells were transfected to express different GFP‐tagged late endosome, MVB, lysosome or autophagosome markers including Rab7 (GFP–Rab7wt), GFP–LC3 and GFP–LAMP1 (green). Cells were double labeled with antibodies to two HBV antigens including anti‐PreS1 of LHBs (A) or an anti‐HBc antibody (red) (B), then analyzed by confocal microscopy. The viral antigens can be found in small putative MVB compartments (Rab7‐positive) but appear most prominent in large autophagosome organelles positive for LAMP1 and LC3. Scale bars: 10 µm. (C,D) Transmission electron microscopy (TEM) of HepG2.2.15 cells showing electron‐dense virus‐like particles (arrows) in MVBs (C) and in a large autophagosome (AP) (D).

Surprisingly, although some HBV‐expressing cells displayed normal spherically shaped MVBs and autophagosomes containing viral proteins (Fig. 1A,B), many possessed large and very long Rab7‐ or LC3‐positive tubules extending from these compartments, which were not found in control cells (Fig. 2A,B; supplementary material Fig. S1E,F). The tubules were also observed in HepG2 cells transfected with 1.3‐fold HBV full genome (Fig. 2C). In many instances, these tubules interconnected to form reticular networks that could also be resolved by TEM (Fig. 2D–F; supplementary material Fig. S1G–J). The viral protein (LHBs) was observed in these tubules (supplementary material Fig. S2A). Importantly, HBV infection also induced the formation of Rab7 tubules in primary human hepatocytes (supplementary material Fig. S2B–E), indicating that this dramatic response was not an aberration that occurs only in a neoplastic cell line.

To test whether other Rab‐centric endocytic pathways are altered by HBV infection, HepG2.2.15 and HepG2 cells were transfected to express GFP‐tagged Rab5 or Rab11. We speculated that changes in these compartments within the infected cells that would represent activation of these Rab proteins similar to that observed for Rab7. No changes in these compartments were observed in infected versus control cells. Because it is thought that MVB functions are associated with the HBV life cycle, we investigated whether there were changes in the distribution of endosomal sorting complexes required for transport (ESCRT) proteins. As well as staining for Hrs, GFP–Tsg101 (ESCRT1), GFP–Vps25 (ESCRT2) or RFP–CHMP6 (ESCRT3) was expressed in these cells but no differences in their distribution was observed (data not shown).

HBV infection increases Rab7 activity and subsequent tubulation of the MVB and autophagosome compartments

To test whether the dramatic morphological changes observed in the late endocytic compartments of HBV‐producing cells (Fig. 2) required the participation of Rab7, both control HepG2 and virus‐expressing HepG2.2.15 cells were treated with Rab7‐specific small interfering RNA (siRNA) to reduce the levels of this small GTPase. The efficiency of Rab7 depletion was good (see Fig. 5A). Indeed, we observed in TEM images very little MVB tubulation following Rab7 depletion (Fig. 3A,B). As assessed by confocal microscopy, Rab7‐depleted cells had significantly less LC3‐positive tubules extending from LC3‐positive organelles than control cells, suggesting that these membrane dynamics required functional Rab7 (Fig. 3C,D).

Fig. 2.

HBV‐producing cells possess highly tubulated MVB‐autophagosome membrane networks containing viral antigens. (A,B) Parental HepG2 cells and HepG2.2.15 cells (A′,A″,B′,B″) were transfected with GFP–Rab7wt (A) or GFP–LC3 (B) and analyzed by confocal microscopy. The MVB and autophagosome compartments are spherical and punctate in the parental cells (A,B), but exceptionally large tubules can be seen extending from Rab7‐ and LC3‐positive compartments (arrows) in the virus‐producing cells. Rab7‐positive tubules (A‴) and LC3‐positive tubules (B‴) in HepG2 and HepG2.2.15 cells were quantified in three independent experiments. The mean±s.e.m. (n = 3) percentage of cells with Rab7 tubules in Rab7‐transfected cells, and the percentage of cells with LC3‐positive tubules in cells with LC3 puncta are shown. (C) HepG2 cells were transfected with 1.3‐fold HBV full genome and GFP–LC3 and analyzed by confocal microscopy. Arrows indicate LC3‐positive tubules. Scale bars: 10 µm. (D,E) TEM images of MVBs in virus‐expressing HepG2.2.15 that extend multiple tubules of uniform diameter (arrows). (F) Quantification (n = 11 cells per condition) of MVB tubules in HepG2 and HepG2.2.15 cells in TEM images reveals a tenfold increase of tubules in virus‐producing cells compared to parental control cells. **P<0.01; ***P<0.001.

Fig. 2.

HBV‐producing cells possess highly tubulated MVB‐autophagosome membrane networks containing viral antigens. (A,B) Parental HepG2 cells and HepG2.2.15 cells (A′,A″,B′,B″) were transfected with GFP–Rab7wt (A) or GFP–LC3 (B) and analyzed by confocal microscopy. The MVB and autophagosome compartments are spherical and punctate in the parental cells (A,B), but exceptionally large tubules can be seen extending from Rab7‐ and LC3‐positive compartments (arrows) in the virus‐producing cells. Rab7‐positive tubules (A‴) and LC3‐positive tubules (B‴) in HepG2 and HepG2.2.15 cells were quantified in three independent experiments. The mean±s.e.m. (n = 3) percentage of cells with Rab7 tubules in Rab7‐transfected cells, and the percentage of cells with LC3‐positive tubules in cells with LC3 puncta are shown. (C) HepG2 cells were transfected with 1.3‐fold HBV full genome and GFP–LC3 and analyzed by confocal microscopy. Arrows indicate LC3‐positive tubules. Scale bars: 10 µm. (D,E) TEM images of MVBs in virus‐expressing HepG2.2.15 that extend multiple tubules of uniform diameter (arrows). (F) Quantification (n = 11 cells per condition) of MVB tubules in HepG2 and HepG2.2.15 cells in TEM images reveals a tenfold increase of tubules in virus‐producing cells compared to parental control cells. **P<0.01; ***P<0.001.

To test whether the observed Rab7‐dependent tubule formation was a result of a specific interaction with its effector protein RILP, which is known to bind active Rab7 and mediate an interaction with microtubule‐based motors (Jordens et al., 2001; van der Kant et al., 2013), we reduced RILP levels in HepG2.2.15 cells by siRNA‐mediated knockdown (supplementary material Fig. S2F,G). This manipulation resulted in a significant reduction in Rab7‐dependent tubule formation and induced a vesiculation of the Rab7 compartment compared to infected control cells (supplementary material Fig. S2H–J). Additionally, expression of exogenous mCherry‐tagged RILP also reduced tubule formation substantially (supplementary material Fig. S2K,L). This response was seen both upon expression of the wild‐type protein, an effect we observe often with an overexpression of tagged proteins, and was accentuated by expression of a tagged Rab7‐binding domain (RBD, amino acids 241–320). Interestingly, expression of an RILP C‐terminal deletion protein (amino acids 1–220, supplementary material Fig. S2K) restored Rab7‐based MVB tubulation in HepG2.2.15 cells substantially to levels seen in cells that had not been transfected with exogenous RILP (supplementary material Fig. S2L), suggesting that expression of a 79 amino acid fragment (amino acids 241–320) makes the protein inert and unrecognizable by the cellular machinery. Taken together, these findings are consistent with the premise that a Rab7–RILP complex mediates HBV‐activated MVB tubulation.

As the tubulation of late endocytic compartments is between three and ten times more prevalent in the virus‐expressing versus control cells (Fig. 2) and is dependent upon Rab7 expression (Fig. 3), we tested whether the HBV is responsible for its activation. To this end, Rab7 activity in HepG2.2.15 cells was compared with that in HepG2 cells using a GST–RILP pulldown assay. Surprisingly, HepG2.2.15 cells had five to eight times higher Rab7 activity than HepG2 control cells (Fig. 4A,B) although there was no difference in total Rab7 levels. Furthermore, HBV from HepG2.2.15 supernatant was added to primary human hepatocytes and also increased Rab7 activity as well as did the secreted viral protein HBsAg, which was detected by an enzyme‐linked immunosorbent assay (ELISA; Fig. 4C,D). To define the HBV component responsible for this dramatic activation, HuH7 cells, which have a higher transfection efficiency than HepG2 cells, were transfected individually with FLAG‐tagged variants of the five distinct proteins encoded by the HBV genome [polymerase, LHBs, HBc, precore, and HBVx protein (HBx)]. Interestingly, the precore protein alone was sufficient to increase Rab7 activity (by fourfold; Fig. 4E,F), although the expression level was significantly lower than that of HBc and HBx and similar to LHBs (supplementary material Fig. S2M). The precore protein is a precursor of HBeAg, and the processed form of HBeAg, in which both N‐terminus and C‐terminus are cleaved (supplementary material Fig. S2N). To further confirm the Rab7‐activating capacity of this viral antigen, the FLAG‐tagged precore protein was expressed in two additional epithelial cell models (Hep3B and HeLa). This protein significantly increased Rab7 activity in both cell lines (Fig. 4G,H), and also the processed form of HBeAg increased Rab7 activity in HuH7 cells (supplementary material Fig. S2N–P). Thus, our data suggests that precore‐ and HBe‐induced Rab7 activation promotes tubulation of late endosomes, MVBs and autophagosomes, which might help regulate virus maturation and subsequent release. The specific knockdown of the precore protein is technically difficult as an siRNA targeting the precore mRNA can also affect the expression of core protein and polymerase.

Fig. 3.

The formation of membranous tubules extending from MVBs and autophagosomes in HBV‐expressing cells is dependent upon Rab7 and facilitates fusion with the lysosome. (A) TEM images of HepG2.2.15 cells treated with non‐targeting (NT) control siRNA or Rab7 siRNA. Representative images of MVBs from NT control and Rab7‐depleted cells are shown. Note the MVB with extending tubules (arrows) in NT siRNA‐treated cells, whereas virus‐producing cells with reduced levels of Rab7 have spherical MVBs with no tubulation. (B) Quantification (n = 12 cells per condition) of MVBs with tubules per cell in control and Rab7‐deficient cells. (C) Rab7 is also required for the dramatic tubulation of the LC3‐positive autophagic compartments. HepG2.2.15 cells were transfected with NT siRNA or Rab7 siRNA, and subsequently transfected to express GFP–LC3. Cells were fixed after 48 h and analyzed with by confocal microscopy. Cells without Rab7 do not show autophagosome tubulation. (D) Percentage of cells with LC3 tubules emanating from LC3 puncta, and numbers of tubules per cell with LC3 puncta are quantified in HepG2.2.15 cells treated with NT siRNA or Rab7 siRNA in three independent experiments. ***P<0.001.

Fig. 3.

The formation of membranous tubules extending from MVBs and autophagosomes in HBV‐expressing cells is dependent upon Rab7 and facilitates fusion with the lysosome. (A) TEM images of HepG2.2.15 cells treated with non‐targeting (NT) control siRNA or Rab7 siRNA. Representative images of MVBs from NT control and Rab7‐depleted cells are shown. Note the MVB with extending tubules (arrows) in NT siRNA‐treated cells, whereas virus‐producing cells with reduced levels of Rab7 have spherical MVBs with no tubulation. (B) Quantification (n = 12 cells per condition) of MVBs with tubules per cell in control and Rab7‐deficient cells. (C) Rab7 is also required for the dramatic tubulation of the LC3‐positive autophagic compartments. HepG2.2.15 cells were transfected with NT siRNA or Rab7 siRNA, and subsequently transfected to express GFP–LC3. Cells were fixed after 48 h and analyzed with by confocal microscopy. Cells without Rab7 do not show autophagosome tubulation. (D) Percentage of cells with LC3 tubules emanating from LC3 puncta, and numbers of tubules per cell with LC3 puncta are quantified in HepG2.2.15 cells treated with NT siRNA or Rab7 siRNA in three independent experiments. ***P<0.001.

Fig. 4.

Rab7 is activated by HBe, a specific gene product of the HBV. (A,B) Cell lysates of HepG2.2.15 cells or parental HepG2 cells were subjected to a GST–RILP pulldown assay to assess Rab7 activity. RILP pulldowns from three independent experiments were subjected to western blot analysis and quantification reveals a marked five to eightfold increase in Rab7 activity in the virus‐producing versus parental cells. (C) As a second model of HBV infection, primary human hepatocytes (PHHs) were incubated with supernatant from HepG2.2.15 cells for 4 h, washed and 7 days later cells were harvested to determine Rab7 activity using a GST–RILP pulldown assay. The HBsAg levels in the supernatant were determined with ELISA. (D) A quantification of active Rab7 as a proportion of total Rab7 for the experiment described in C, shows a greater than twofold increase in Rab7 activity above infected control cells. (E) The precore protein, a precursor of HBeAg, activates Rab7. Lysates of HuH7 cells, previously transfected to express five distinct FLAG‐tagged HBV proteins (polymerase, LHBs, HBc, precore and HBx), were subjected to GST–RILP pulldown assays to test for the component that induces the highest Rab7 activity. (F) Quantification of active Rab7 levels in three independent experiments as described in E revealed that cells expressing the viral precore protein activated four to five times more Rab7 than the other expressed viral components. (G,H) To test whether the precore protein might activate Rab7 in other epithelial cells, Hep3B or HeLa cells were also transfected with FLAG‐tagged precore protein, lysed and subjected to a GST–RILP pulldown assays (G). A quantification of active Rab7 as a proportion of total Rab7 in three independent experiments indicates that the precore protein alone markedly activates Rab7 in other epithelial cells, including non‐hepatocytes such as HeLa (H). Pol, polymerase; PreC, precore. *P<0.05.

Fig. 4.

Rab7 is activated by HBe, a specific gene product of the HBV. (A,B) Cell lysates of HepG2.2.15 cells or parental HepG2 cells were subjected to a GST–RILP pulldown assay to assess Rab7 activity. RILP pulldowns from three independent experiments were subjected to western blot analysis and quantification reveals a marked five to eightfold increase in Rab7 activity in the virus‐producing versus parental cells. (C) As a second model of HBV infection, primary human hepatocytes (PHHs) were incubated with supernatant from HepG2.2.15 cells for 4 h, washed and 7 days later cells were harvested to determine Rab7 activity using a GST–RILP pulldown assay. The HBsAg levels in the supernatant were determined with ELISA. (D) A quantification of active Rab7 as a proportion of total Rab7 for the experiment described in C, shows a greater than twofold increase in Rab7 activity above infected control cells. (E) The precore protein, a precursor of HBeAg, activates Rab7. Lysates of HuH7 cells, previously transfected to express five distinct FLAG‐tagged HBV proteins (polymerase, LHBs, HBc, precore and HBx), were subjected to GST–RILP pulldown assays to test for the component that induces the highest Rab7 activity. (F) Quantification of active Rab7 levels in three independent experiments as described in E revealed that cells expressing the viral precore protein activated four to five times more Rab7 than the other expressed viral components. (G,H) To test whether the precore protein might activate Rab7 in other epithelial cells, Hep3B or HeLa cells were also transfected with FLAG‐tagged precore protein, lysed and subjected to a GST–RILP pulldown assays (G). A quantification of active Rab7 as a proportion of total Rab7 in three independent experiments indicates that the precore protein alone markedly activates Rab7 in other epithelial cells, including non‐hepatocytes such as HeLa (H). Pol, polymerase; PreC, precore. *P<0.05.

Rab7 depletion increases HBV secretion

As HBV infection results in a profound remodeling of the Rab7‐associated MVB and autophagosome compartments in cells it seemed likely that these changes could affect HBV assembly, intracellular trafficking, and secretion from cells. To test this, Rab7 was depleted in HepG2.2.15 cells by siRNA and the levels of internal viral proteins were assayed by western blot analysis and the level of secreted virus was assayed by real‐time PCR. Surprisingly, we observed that Rab7 depletion induced a threefold increase in the intracellular accumulation of LHBs (Fig. 5A,B) and also significantly increased the release of HBV DNA into the supernatant (Fig. 5C). To confirm that the increase in HBV DNA released from Rab7‐depleted cells represents intact enveloped virus, HepG2.2.15 cells were transfected with FLAG–HBs and the supernatant was subjected to an immunoprecipitation with anti‐FLAG antibody and then subjected to PCR for the HBV genome. The rationale for this approach was that the tagged HBs could associate with and subsequently precipitate the viral genome for detection only if packaged into a virion. As shown in Fig. 5D, the levels of HBV DNA post‐immunoprecipitation were also increased in the supernatant from Rab7‐depleted cells, an indication that Rab7 depletion increases the secretion of enveloped HBV particles. In control experiments, the expression of FLAG‐tagged mock protein or an addition of control antibody did not increase the HBV DNA levels after immunoprecipitation. There was no significant difference in the amount of HBsAg in the supernatant from cells with or without Rab7 depletion (data not shown), suggesting that the secretion pathways of HBV particles and HBsAg subviral particles (SVPs) are different, as reported previously (Garcia et al., 2009), and Rab7 does not affect the secretion pathway of SVPs. The expression of a GTPase‐defective Rab7 mutant (Rab7T22N) also increased the amount of intracellular LHBs and released viral genome, as with the Rab7 knockdown (Fig. 5E,F). Additionally, the expression of mCherry–RILP reduced the secretion of HBV DNA in the supernatant but the effect was not seen after Rab7 knockdown (supplementary material Fig. S3A,B).

Fig. 5.

Inhibiting Rab7 function induces a marked increase in HBV secretion. (A) HepG2.2.15 cells were transfected with non‐targeting (NT) control siRNA or Rab7 siRNA, and 3 days after the transfection, the levels of LHBs in the cells were determined by western blot analysis. Note that Rab7 depletion substantially increases the amount of intracellular LHBs. (B) Quantification of LHBs in five independent experiments as described in A confirm a greater than threefold accumulation of LHBs protein in Rab7‐deficient cells. (C) As a measure of viral secretion, total HBV DNA recovered from the supernatant of cells with or without Rab7 depletion was determined by real‐time PCR in five independent experiments and showed a substantial increase (>threefold) in the amount of viral genome released. (D) Rab7 depletion increases the release of intact enveloped virion. Rab7 was knocked down in HepG2.2.15 cells, followed by transfection of FLAG–HBs 24 h later. Two days following the transfection procedure, enveloped HBV in the supernatant was precipitated with the anti‐FLAG antibody and the levels of HBV DNA from the precipitates were determined by real‐time PCR. Three independent experiments revealed a significant increase in the release of enveloped HBV from the cells with reduced Rab7 levels. (E,F) Inactive Rab7 increases HBV secretion. HepG2.2.15 cells were transfected twice with GFP–Rab7wt or the GTPase‐defective GFP–Rab7T22N mutant, and the levels of LHBs were determined by western blot analysis (E) and total HBV DNA in the supernatant was determined with real‐time PCR (F). Quantification of three independent experiments are shown and indicate an increase in HBV secretion in the mutant compared to the wild‐type‐expressing cells, similar to that observed in the Rab7‐depleted cells. *P<0.05; **P<0.01; ***P<0.001.

Fig. 5.

Inhibiting Rab7 function induces a marked increase in HBV secretion. (A) HepG2.2.15 cells were transfected with non‐targeting (NT) control siRNA or Rab7 siRNA, and 3 days after the transfection, the levels of LHBs in the cells were determined by western blot analysis. Note that Rab7 depletion substantially increases the amount of intracellular LHBs. (B) Quantification of LHBs in five independent experiments as described in A confirm a greater than threefold accumulation of LHBs protein in Rab7‐deficient cells. (C) As a measure of viral secretion, total HBV DNA recovered from the supernatant of cells with or without Rab7 depletion was determined by real‐time PCR in five independent experiments and showed a substantial increase (>threefold) in the amount of viral genome released. (D) Rab7 depletion increases the release of intact enveloped virion. Rab7 was knocked down in HepG2.2.15 cells, followed by transfection of FLAG–HBs 24 h later. Two days following the transfection procedure, enveloped HBV in the supernatant was precipitated with the anti‐FLAG antibody and the levels of HBV DNA from the precipitates were determined by real‐time PCR. Three independent experiments revealed a significant increase in the release of enveloped HBV from the cells with reduced Rab7 levels. (E,F) Inactive Rab7 increases HBV secretion. HepG2.2.15 cells were transfected twice with GFP–Rab7wt or the GTPase‐defective GFP–Rab7T22N mutant, and the levels of LHBs were determined by western blot analysis (E) and total HBV DNA in the supernatant was determined with real‐time PCR (F). Quantification of three independent experiments are shown and indicate an increase in HBV secretion in the mutant compared to the wild‐type‐expressing cells, similar to that observed in the Rab7‐depleted cells. *P<0.05; **P<0.01; ***P<0.001.

To further define the role of Rab7 function in the HBV life cycle, ‘rescue’ experiments were performed in which GFP–Rab7wt or GFP–Rab7T22N were re‐expressed in the Rab7‐depleted cells. As expected, we observed that GFP–Rab7wt re‐expression, but not that of GFP–Rab7T22N, restored the LHBs level (supplementary material Fig. S3C,D) as well as the colocalization of LHBs and LAMP1 (supplementary material Fig. S3E,F). In addition, the reduced number of LC3‐positive tubules seen after Rab7 depletion were restored by the expression of FLAG–Rab7wt (supplementary material Fig. S3G–I). These data provide additional support for the concept that the effects of the Rab7 depletion on HBV production and localization were not due to off‐target effects. More importantly, the viruses secreted from the Rab7‐depleted cells appear to represent infectious particles because HuS‐E/2 cells, which are susceptible to HBV infection (Huang et al., 2012), show equal infectivity in response to virus collected from either knockdown or control cells (supplementary material Fig. S4A–C). A successful infection was confirmed by the gradual increase of HBV DNA in the supernatant from HuS‐E/2 cells (supplementary material Fig. S4A).

From the findings described above, we hypothesized that the effect of decreased Rab7 levels leading to an increase in both internal and secreted virus could be a result of altered trafficking to the degradative lysosome. To test whether this transport occurs in live cells, we analyzed HepG2.2.15 cells expressing GFP–LC3 and mCherry–LAMP1 by confocal microscopy. As depicted in Fig. 6A, time‐lapse images revealed that LAMP1 puncta were tethered to and then fused with LC3‐positive tubules or, alternatively, were pulled towards an autophagosome. Therefore, it was considered that the Rab7‐dependent tubules induced by HBV might facilitate the lysosomal degradation of virus in MVBs or autophagosomes. Accordingly, we tested for changes in the distribution of viral proteins and lysosomes in virus‐producing cells in the context of Rab7 depletion. Knockdown of Rab7 in these cells induced a significant (70%) decrease in the localization of LHBs with LAMP1 compartments (Fig. 6B,C), suggesting that Rab7 facilitates the transport to and subsequent degradation of HBV in lysosomes. In addition and consistent with previous observations (Vanlandingham and Ceresa, 2009), significantly more MVBs were found in Rab7‐depleted cells when assessed by TEM (Fig. 6D–F), suggesting an induced defect in MVB–lysosome fusion. In support of this premise, we observed enlarged MVBs in Rab7‐depleted cells that were not seen in control cells (supplementary material Fig. S4D,E).

Fig. 6.

Rab7‐dependent tubules facilitate the fusion of MVBs or autophagosomes with lysosomes. (A) LC3‐positive tubules in HBV‐producing cells interact with LAMP1‐positive lysosomes. HepG2.2.15 cells were transfected with GFP–LC3 and mCherry–LAMP1, and time‐lapse confocal images were obtained over 20–30 min. Two representative image series show a physical interaction between the autophagosomes and lysosomes (arrows). (B,C) Altering Rab7 function reduces the transport of viral proteins from the MVB or autophagosome to the lysosome. HepG2.2.15 cells were treated with non‐targeting (NT) siRNA or Rab7 siRNA followed by transfection of mCherry–LAMP1, and then stained with an anti‐PreS1 antibody labeling LHBs. Cells with impaired Rab7 function showed a marked reduction of transported HBV proteins to the LAMP1‐positive lysosomal compartment. Scale bars: 10 µm. The graph represents three independent experiments where the numbers of LHBs‐ and LAMP1‐positive vesicles as a proportion of the total LHBs‐positive vesicles were calculated. (D,E) TEM images of HepG2.2.15 cells treated with NT siRNA (D) or Rab7 siRNA (E) showing a substantial increase in the number of accumulated MVBs. (F) Quantification (n = 12 cells per condition) of MVBs per cell with NT siRNA or Rab7 siRNA. ***P<0.001.

Fig. 6.

Rab7‐dependent tubules facilitate the fusion of MVBs or autophagosomes with lysosomes. (A) LC3‐positive tubules in HBV‐producing cells interact with LAMP1‐positive lysosomes. HepG2.2.15 cells were transfected with GFP–LC3 and mCherry–LAMP1, and time‐lapse confocal images were obtained over 20–30 min. Two representative image series show a physical interaction between the autophagosomes and lysosomes (arrows). (B,C) Altering Rab7 function reduces the transport of viral proteins from the MVB or autophagosome to the lysosome. HepG2.2.15 cells were treated with non‐targeting (NT) siRNA or Rab7 siRNA followed by transfection of mCherry–LAMP1, and then stained with an anti‐PreS1 antibody labeling LHBs. Cells with impaired Rab7 function showed a marked reduction of transported HBV proteins to the LAMP1‐positive lysosomal compartment. Scale bars: 10 µm. The graph represents three independent experiments where the numbers of LHBs‐ and LAMP1‐positive vesicles as a proportion of the total LHBs‐positive vesicles were calculated. (D,E) TEM images of HepG2.2.15 cells treated with NT siRNA (D) or Rab7 siRNA (E) showing a substantial increase in the number of accumulated MVBs. (F) Quantification (n = 12 cells per condition) of MVBs per cell with NT siRNA or Rab7 siRNA. ***P<0.001.

To investigate the effect of HBV on endocytic uptake and trafficking of non‐viral endogenous receptors, we compared the kinetics of the epidermal growth factor receptor (EGFR) following stimulation with EGF ligand in HepG2 versus the infected HepG2.2.15 cells as described previously (Schroeder et al., 2010). Although HepG2.2.15 cells exhibited markedly higher levels of EGFR to begin with, the kinetics of degradation up to 6 h appeared very similar. Furthermore, we observed little difference in the uptake and trafficking of rhodamine‐tagged transferrin (data not shown).

Inhibiting lysosome function increases HBV secretion

Because Rab7 appears to be mediating the transfer of nascent HBV from MVBs to the lysosome for potential degradation, we tested whether treating cells with CQ, which disrupts lysosome function by preventing acidification, might also increase viral secretion and therefore mimic the effects of the Rab7 knockdown. HepG2.2.15 cells were treated with 0–50 µM CQ for 24 h and the levels of LHBs in the cells and HBV DNA in the supernatant were assayed. As predicted, both were increased significantly after CQ treatment in a dose‐dependent manner (Fig. 7A–C). In addition, another lysosome inhibitor NH4Cl produced similar results in two independent experiments (supplementary material Fig. S4F,G). This indicates that a substantial percentage of HBV is degraded in lysosomes under normal conditions. When the virus‐containing cell supernatant was collected from the drug‐treated cells and added to HuS‐E/2 cells (the CQ concentration was diluted 300 times beyond its working concentration), we did not see a significant difference in the infectivity compared to control cells (data not shown), similar to our observations using virus‐containing supernatant from Rab7‐depleted cells (supplementary material Fig. S4A–C). Most importantly, the CQ‐treated cells no longer exhibited a Rab7‐depletion effect on the intracellular levels of LHBs (Fig. 7D,E), supporting the concept that Rab7 mediates the transport of HBV to the lysosome to reduce viral production and secretion.

Fig. 7.

Inhibition of lysosomal function prevents the Rab7‐dependent reduction of HBV. (A,B) HBV levels in cells are reduced by lysosomal degradation. HepG2.2.15 cells were treated with the lysosome inhibitor chloroquine (CQ) at various concentrations for 24 h (A) and the levels of LHBs were determined and quantified by western blot analysis (B) from three independent experiments. A substantial, seven to eightfold retention of viral protein was observed in cells upon disruption of lysosomal function. (C) Total HBV DNA in the culture supernatant of HepG2.2.15 cells treated with CQ was determined with real‐time PCR (n = 3) and reveals a three to fourfold increase in viral secretion. (D,E) Rab7 supports lysosome‐mediated degradation of HBV. HepG2.2.15 cells were transfected with Rab7 or NT siRNA prior to treatment with 25 µM CQ. 24 h following treatment, LHBs levels were determined and quantified by western blot analysis (D) in three independent experiments (E). The siRNA‐mediated reduction of Rab7 resulted in a twofold increase in total cellular LHBs protein, an effect that was negated by CQ treatment. *P<0.05; **P<0.01.

Fig. 7.

Inhibition of lysosomal function prevents the Rab7‐dependent reduction of HBV. (A,B) HBV levels in cells are reduced by lysosomal degradation. HepG2.2.15 cells were treated with the lysosome inhibitor chloroquine (CQ) at various concentrations for 24 h (A) and the levels of LHBs were determined and quantified by western blot analysis (B) from three independent experiments. A substantial, seven to eightfold retention of viral protein was observed in cells upon disruption of lysosomal function. (C) Total HBV DNA in the culture supernatant of HepG2.2.15 cells treated with CQ was determined with real‐time PCR (n = 3) and reveals a three to fourfold increase in viral secretion. (D,E) Rab7 supports lysosome‐mediated degradation of HBV. HepG2.2.15 cells were transfected with Rab7 or NT siRNA prior to treatment with 25 µM CQ. 24 h following treatment, LHBs levels were determined and quantified by western blot analysis (D) in three independent experiments (E). The siRNA‐mediated reduction of Rab7 resulted in a twofold increase in total cellular LHBs protein, an effect that was negated by CQ treatment. *P<0.05; **P<0.01.

In this study, we provide novel findings implicating the small GTPase Rab7 as a central regulator of HBV transport and secretion. We find that assembled virus accumulates in Rab7‐positive MVB and autophagosome compartments (Fig. 1), which then become markedly altered and tubulated upon infection of hepatocytes (Fig. 2). Equally surprising was the finding that HBV significantly enhances Rab7 activity through the expression of the precore or HBe protein encoded by its own invasive genome (Fig. 4). In this context, Rab7‐dependent activation of the late endosomal pathway (Fig. 3) appears to facilitate the interaction with, and potentially exchange of contents between (Fig. 6), the MVB, autophagosome and lysosome (Figs 6,7), leading to an increase in HBV degradation and a subsequent attenuation of viral shedding (Fig. 5). Fig. 8 illustrates our main concept, demonstrating that the HBV infection promotes the formation of an interconnecting reticular network between MVBs, autophagosomes and lysosomes driven by the activation of Rab7. Although tubules extending from MVBs and autophagosomes have been reported previously (Cooney et al., 2002; Cortese et al., 2013; Gao et al., 2010), those induced by HBV in the current study are much more prominent.

Fig. 8.

Model depicting the role of Rab7 in the regulation of HBV secretion. (A) In uninfected cells with basal Rab7 activity, MVB and autophagosome (AP) tubules are less prominent resulting in normal exocytosis. (B) The viral HBe protein within infected cells activates Rab7, inducing the formation of an interconnected tubular‐reticular network of MVBs, autophagosomes and lysosomes along microtubules. This network promotes the transfer of virus between the various compartments, resulting in the degradation of nascent virus and reduced secretion. (C) In Rab7‐depleted cells, the interaction between these compartments is substantially reduced, leading to an accumulation of MVBs and a subsequent increase the levels of intracellular and released virus, a result of reduced viral transport to the lysosome.

Fig. 8.

Model depicting the role of Rab7 in the regulation of HBV secretion. (A) In uninfected cells with basal Rab7 activity, MVB and autophagosome (AP) tubules are less prominent resulting in normal exocytosis. (B) The viral HBe protein within infected cells activates Rab7, inducing the formation of an interconnected tubular‐reticular network of MVBs, autophagosomes and lysosomes along microtubules. This network promotes the transfer of virus between the various compartments, resulting in the degradation of nascent virus and reduced secretion. (C) In Rab7‐depleted cells, the interaction between these compartments is substantially reduced, leading to an accumulation of MVBs and a subsequent increase the levels of intracellular and released virus, a result of reduced viral transport to the lysosome.

Rab7 is a key trafficking regulator involved in multiple post‐endocytic processes including transport between late endosomes/MVBs, autophagosomes, lysosomes and other lysosome‐related organelles (Wang et al., 2011). Activated Rab7 on the endosomal membrane binds to RILP, a well‐characterized effector of Rab7, together forming a complex with p150Glued (also known as DCTN1) and dynein–dynactin to drive the transport of endosomes to the minus‐end of microtubules (Jordens et al., 2001). Additionally, Rab7 is known to associate with the FYVE coiled coil domain protein (FYCO1) and the molecular motor kinesin to participate in the microtubule plus‐end‐directed transport of endosomes (Pankiv et al., 2010). The activation of this complex might promote the formation of tubular extensions bidirectionally along microtubules, as discussed previously (Harrison et al., 2003), and, as observed by our electron micrographs (Fig. 3A; supplementary material Fig. S1J). Although many tubules were found to extend from MVBs and autophagosomes in the HBV‐producing cells (Fig. 2), we observed no alterations in the lysosomal structure (data not shown), suggesting that Rab7 specifically acted in the MVB and autophagosome compartments.

Currently, the mechanism by which the precore or HBe protein activates Rab7 is unclear as we have been unable to detect any interactions between the viral protein and Rab7 biochemically (data not shown). It is possible, if not likely, that the HBe protein could interact with either guanine‐nucleotide‐exchange factors (GEFs) or GTPase‐activating proteins (GAPs) specific for Rab7 (Wang et al., 2011). Interaction with these regulatory proteins could activate Rab7 by enhancing the function of a GEF, or in contrast, inhibiting the function of a GAP. Studies to test for interactions between HBe and these accessory factors are ongoing.

A central finding of this study indicates that the HBV itself activates Rab7 to promote exchange between MVBs, autophagosomes and the lysosome (Fig. 6) resulting in reduced viral production. The marked intracellular accumulation of viral proteins accompanied by the increase in viral secretion following Rab7 knockdown (Fig. 5) or inhibition of lysosome function (Fig. 7) supports this concept. Some possibilities exist to explain how the virus would benefit from activating the pathway that would direct it into the lysosome for eventual destruction. First, processing of surface proteins by lysosomal enzymes might aid in viral ‘maturation’ and increase infectivity, or, alternatively, decrease viral recognition by neutralizing antibodies in the blood. To test these concepts, we compared the infectivity of virus released from cells with or without Rab7 depletion, as well as with or without CQ treatment; however, no differences in infectivity were observed (supplementary material Fig. S4A–C). Second, and perhaps more relevant, is the fact that HBV is considered a ‘stealth virus’ that is able to escape from the front line of host defenses (Wieland and Chisari, 2005). HBe, which is not required for viral replication, is considered to have a role in the establishment of chronic infection (Milich and Liang, 2003). It has been shown that HBe modulates innate immune signal transduction pathways through interaction with and targeting of the signaling pathways mediated by Toll‐like receptors (Lang et al., 2011). As a result of attenuated immune responses, the viral load increases greatly in the early phase of chronic infection (Liang, 2009). We hypothesize that HBV might put a brake on its own secretion through HBe‐mediated Rab7 activation to reduce the immune response. In support of this hypothesis, it has been shown that HBe‐negative strains have high replication capacity in vitro (Inoue et al., 2011; Ozasa et al., 2006) and that they can cause vigorous immune responses resulting in fulminant hepatitis (Milich and Liang, 2003).

An alternative explanation as to why we observed this Rab7 activation is that the activation of a Rab7‐mediated viral degradation pathway rather than representing a host defense mechanism – that is, hepatocytes respond to the expression of the HBe antigen by grossly activating the tubulation and fusion of MVBs and autophagosomes with the lysosome. Such membrane remodeling events could be part of an autophagy‐mediated clearance of invading pathogens (xenophagy), a well‐established cellular defense mechanism (Levine, 2005).

Finally, it is important to note that the specific role of Rab7 described here might represent just one of several functions in the HBV life cycle. A recent paper has shown that the early entry stages of HBV infection in HepaRG cells depend on both Rab5 and Rab7 (Macovei et al., 2013). The HepG2.2.15 cell model used in our current study stably expresses HBV and is not susceptible to further infection because it expresses very low levels of the putative HBV receptor, the sodium taurocholate cotransporting polypeptide (NTCP) (Yan et al., 2012). Therefore, HepG2.2.15 cells provide a useful model to study the production and release of the virus rather than infection. Thus, Rab7 activation by the HBe protein might also increase the efficiency of the early stages of infection.

It is clear from this and other studies implicating the endosomal pathways in HBV infection that a more complete understanding of how this virus usurps the vesicle trafficking machinery from the hepatocyte to suit its own ends will be a complex but rewarding challenge. Additional regulatory Rab GTPases, vesicle coat and adaptor proteins, as well as fission enzymes, are likely to participate in the HBV life cycle and thus will provide useful drug targets for future therapy.

Plasmids and siRNA

To obtain FLAG‐tagged HBV individual protein constructs, individual DNA sequences specific for each protein were amplified from a total DNA extracted from the culture supernatant of HepG2.2.15 cells. Nucleotides [nt, the numbers are in accordance with a genotype D HBV sequence of 3182 nt from HepG2.2.15 (accession number U95551)] 2307–3182 and 1–1623, 2847–3182 and 1–835, 155–835, 1899–2453, 1814–2453, and 1374–1840 were amplified for FLAG–polymerase, FLAG–LHBs, FLAG–HBs, FLAG–HBc, FLAG–precore and FLAG–HBx, respectively. These PCR products were cloned into pcDNA3 (Invitrogen, Carlsbad, CA) modified to have a FLAG sequence upstream of the multiple‐cloning site. 1.3‐fold wild‐type HBV genome (nt 1051–3215 and 1–1953, which is 1.3-fold longer than a circular HBV genome) of genotype B, which was obtained from an acute hepatitis patient, was described previously (Inoue et al., 2011). GFP–Rab7wt was as described previously (Schroeder et al., 2012) and GFP–Rab7T22N was kindly provided by Dr Bruce Horazdovsky (Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN). FLAG–Rab7wt was made from a PCR product that was amplified from GFP–Rab7wt. GST–RILP was kindly provided by Dr Cecilia Bucci (Universita del Salento, Italy) and mCherry–RILP was provided by Dr Barbara Schroeder (Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN) and obtained by cloning the RILP sequence into the HindIII/XbaI sites of pmCherry‐C3 (Takara Bio, Shiga, Japan). GFP–LAMP1 and mCherry–LAMP1 were made as described previously (Schulze et al., 2013). siRNA targeting human Rab7, whose sequence was described previously (Jäger et al., 2004), siRNA targeting human RILP (5′‐GAUCAAGGCCAAGAUGUUAUU‐3′) and a non‐targeting (NT) control siRNA were purchased from Dharmacon (Thermo Fisher Scientific, Lafayette, CO).

Antibodies and other reagents

Anti‐PreS1 and anti‐GST antibodies were purchased from Santa Cruz Biotechnology (Santa Cruz, CA). Anti‐HBc was purchased from Abcam (Cambridge, UK). Antibodies against Rab7, FLAG (monoclonal) and actin, as well as chloroquine (CQ), were purchased from Sigma‐Aldrich (St. Louis, MO). Anti‐FLAG (polyclonal) and anti‐Myc (monoclonal and polyclonal) antibodies were purchased from Cell Signaling (Danvers, MA). Anti‐Hrs antibody was purchased from Bethyl Laboratories (Montgomery, TX). Anti‐LC3 was purchased from Novus Biologicals (Littleton, CO).

Cell culture and transfection

The HBV‐expressing stable cell line HepG2.2.15, which was kindly provided by Dr Andrea Cuconati (Institute of Hepatitis and Virus Research, Doylestown, PA), and the parental human hepatoma cell line HepG2 were incubated in RPMI 1640 with L‐glutamine (Corning Cellgro, Manassas, VA) supplemented with 10% fetal bovine serum (FBS), 50 U/ml penicillin and 50 µg/ml streptomycin. HepG2.2.15 and HepG2 cells were seeded onto coverslips or plates coated with poly‐L‐lysine (Sigma‐Aldrich) as described previously (Abdulkarim et al., 2003). Cells were transiently transfected with Lipofectamine Plus (Invitrogen) or Lipofectamine 2000 (Invitrogen) according to the manufacturer's instructions. HepG2.2.15 cells were transfected twice in a 24 h interval to increase the transfection efficiency. Transfection of HepG2.2.15 cells with siRNA was performed using RNAiMAX (Invitrogen) following the standard protocol. The re‐expression was performed as above 24 h after the siRNA transfection. HeLa and the human hepatoma cell lines HuH7 and Hep3B were incubated in MEM (Corning cellgro), supplemented with 10% FBS, 1.5 g/l sodium bicarbonate, 1× nonessential amino acids, 1 mM sodium pyrophosphate, 50 U/ml penicillin and 50 µg/ml streptomycin. An immortalized human primary hepatocyte cell line HuS‐E/2, which was kindly provided by Dr Makoto Hijikata (Kyoto University, Japan), was grown as described previously (Aly et al., 2007). HBV infection experiments with HuS‐E/2 cells were performed as described previously (Huang et al., 2012). Primary human hepatocytes (PHHs) were from Yecuris (Tualatin, OR) and maintained in DMEM with high glucose and L‐glutamine, supplemented with 10% FBS and 50 U/ml penicillin and 50 µg/ml streptomycin.

Western blot analysis

Cells were lysed in RIPA buffer [150 mM NaCl, 1% NP‐40, 0.5% deoxycholic acid, 0.1% SDS, 50 mM Tris‐HCl pH 8.0 and complete protease inhibitors (Roche Diagnostics, Indianapolis, IN)]. 30 µg of soluble proteins were resolved by SDS‐PAGE, and transferred onto a PVDF membrane. After blocking with 10% milk in PBS, the membrane was incubated with primary antibodies at room temperature for 2 h, and incubated with secondary antibodies for 1 h. The signals were detected with SuperSignal West Pico Chemiluminescent Substrate (Thermo Fisher Scientific).

GST–RILP pulldown

GST–RILP was transformed into Escherichia coli BL21 cells and 4 ml of an overnight culture was cultured further in 200 ml LB to an optical density (OD) at 600 nm of 0.6–0.8. After the addition of isopropyl β‐D‐1‐thiogalactopyranoside (IPTG, final concentration of 1 mM), it was incubated at room temperature for 3–4 h. The culture was spun down, and the collected bacteria were sonicated in PBS on ice. They were rotated at 4°C for 30 min with 1% Triton X‐100 and GST–RILP protein was purified using glutathione–Sepharose‐4B beads (Amersham‐Pharmacia Biotech, GE Healthcare Bio‐Sciences, Piscataway, NJ) according to the manufacturer's instructions. The beads and 0.25 mg of cell lysate to be analyzed were rocked in 600 µl of Bing buffer (25 mM Tris-HCI pH 7.4, 100 mM NaCI, 1 mM DTT, 1% NP-40, 2mM Na3VO4, 15 mM NaF, 0.1 mM EDTA and complete protease inhibitors) at 4°C for 1–2 h. The levels of active Rab7 binding to GST–RILP were quantified by western blot analysis.

Real‐time PCR and ELISA

Total DNA was extracted with a QIAamp DNA Blood Mini Kit (QIAGEN, Hilden, Germany) from 100 µl of the culture supernatant, and 2 µl of the extracted DNA solution was subjected to real‐time PCR using a LightCycler 480 system (Roche Diagnostics) with SYBR Green I Master (Roche Diagnostics), primers HBXF11 (5′‐ATGGCTGCTAGGCTGTGCTG‐3′) and HBXR4 (5′‐GTCCGCGTAAAGAGAGGTGC‐3′). The HBsAg levels in the culture supernatant were quantified with Hepatitis B Surface Antigen ELISA Kit (MyBioSource, San Diego, CA).

Immunoprecipitation of viral particles

HepG2.2.15 cells were transfected with FLAG–HBs, and 2 days following transfection, enveloped HBV particles in the supernatant were precipitated using the anti‐FLAG antibody. The levels of HBV DNA from the precipitates were determined by real‐time quantitative PCR.

Immunofluorescence and transmission electron microscopy

Fluorescence micrographs were acquired using a Zeiss Axio Observer.D1 microscope (Carl Zeiss, Oberkochen, Germany) with a 63× objective, an Orca II camera (Hamamatsu Photonics, Hamamatsu, Japan), and iVision software (BioVision Technologies, Exton, PA), or an LSM780 confocal microscope (Carl Zeiss) with a 63× objective and ZEN software (Carl Zeiss). Time‐lapse images of live cells were taken using the confocal microscope every 30 s on a heated chamber set at 37°C and under 5% CO2. Transmission electron microscopy (TEM) micrographs were acquired as previously described (Henley et al., 1998) with a JEOL 1200 electron microscope (JEOL Ltd, Tokyo, Japan). Images were adjusted with Adobe Photoshop software (Adobe, San Jose, CA).

Statistical analysis

Results of quantitative variables are expressed as mean±s.e.m. Statistical comparisons were made by using a Student's t‐test unless otherwise indicated and P<0.05 was considered significant.

The authors would like to thank members of the McNiven laboratory for their contributions, especially Barbara Schroeder and Ryan Schulze for critical readings of the manuscript.

Author contributions

J.I. designed and performed experiments, analyzed data and wrote the paper; E.W.K. performed microscopy experiments; J.C. assisted with protein assays; H.C. performed experiments of RILP knockdown and expression, EGFR trafficking and transferrin trafficking; M.N. assisted with HBV infection and quantification; M.A.M. designed experiments and wrote the paper.

Funding

This study was supported by the Japan Society for the Promotion of Science (to J.I.); the Mochida Memorial Foundation for Medical and Pharmaceutical Research (to J.I.); and by the National Institute of Diabetes and Digestive and Kidney Diseases [grant number DK44650 to M.A.M.]. Deposited in PMC for release after 12 months.

Abdulkarim
AS
,
Cao
H
,
Huang
B
,
McNiven
MA
2003
The large GTPase dynamin is required for hepatitis B virus protein secretion from hepatocytes.
J. Hepatol.
38
76
83
Aly
HH
,
Watashi
K
,
Hijikata
M
,
Kaneko
H
,
Takada
Y
,
Egawa
H
,
Uemoto
S
,
Shimotohno
K
2007
Serum-derived hepatitis C virus infectivity in interferon regulatory factor-7-suppressed human primary hepatocytes.
J. Hepatol.
46
26
36
Berg
TO
,
Fengsrud
M
,
Strømhaug
PE
,
Berg
T
,
Seglen
PO
1998
Isolation and characterization of rat liver amphisomes. Evidence for fusion of autophagosomes with both early and late endosomes.
J. Biol. Chem.
273
21883
21892
Bruss
V
2007
Hepatitis B virus morphogenesis.
World J. Gastroenterol.
13
65
73
Bucci
C
,
Thomsen
P
,
Nicoziani
P
,
McCarthy
J
,
van Deurs
B
2000
Rab7: a key to lysosome biogenesis.
Mol. Biol. Cell
11
467
480
Cantalupo
G
,
Alifano
P
,
Roberti
V
,
Bruni
CB
,
Bucci
C
2001
Rab-interacting lysosomal protein (RILP): the Rab7 effector required for transport to lysosomes.
EMBO J.
20
683
693
Cooney
JR
,
Hurlburt
JL
,
Selig
DK
,
Harris
KM
,
Fiala
JC
2002
Endosomal compartments serve multiple hippocampal dendritic spines from a widespread rather than a local store of recycling membrane.
J. Neurosci.
22
2215
2224
Cortese
K
,
Howes
MT
,
Lundmark
R
,
Tagliatti
E
,
Bagnato
P
,
Petrelli
A
,
Bono
M
,
McMahon
HT
,
Parton
RG
,
Tacchetti
C
2013
The HSP90 inhibitor geldanamycin perturbs endosomal structure and drives recycling ErbB2 and transferrin to modified MVBs/lysosomal compartments.
Mol. Biol. Cell
24
129
144
Eble
BE
,
MacRae
DR
,
Lingappa
VR
,
Ganem
D
1987
Multiple topogenic sequences determine the transmembrane orientation of the hepatitis B surface antigen.
Mol. Cell. Biol.
7
3591
3601
Fader
CM
,
Colombo
MI
2009
Autophagy and multivesicular bodies: two closely related partners.
Cell Death Differ.
16
70
78
Fader
CM
,
Sánchez
D
,
Furlán
M
,
Colombo
MI
2008
Induction of autophagy promotes fusion of multivesicular bodies with autophagic vacuoles in k562 cells.
Traffic
9
230
250
Gao
W
,
Kang
JH
,
Liao
Y
,
Ding
WX
,
Gambotto
AA
,
Watkins
SC
,
Liu
YJ
,
Stolz
DB
,
Yin
XM
2010
Biochemical isolation and characterization of the tubulovesicular LC3-positive autophagosomal compartment.
J. Biol. Chem.
285
1371
1383
Garcia
T
,
Li
J
,
Sureau
C
,
Ito
K
,
Qin
Y
,
Wands
J
,
Tong
S
2009
Drastic reduction in the production of subviral particles does not impair hepatitis B virus virion secretion.
J. Virol.
83
11152
11165
Hanson
PI
,
Cashikar
A
2012
Multivesicular body morphogenesis.
Annu. Rev. Cell Dev. Biol.
28
337
362
Harrison
RE
,
Bucci
C
,
Vieira
OV
,
Schroer
TA
,
Grinstein
S
2003
Phagosomes fuse with late endosomes and/or lysosomes by extension of membrane protrusions along microtubules: role of Rab7 and RILP.
Mol. Cell. Biol.
23
6494
6506
Henley
JR
,
Krueger
EW
,
Oswald
BJ
,
McNiven
MA
1998
Dynamin-mediated internalization of caveolae.
J. Cell Biol.
141
85
99
Huang
HC
,
Chen
CC
,
Chang
WC
,
Tao
MH
,
Huang
C
2012
Entry of hepatitis B virus into immortalized human primary hepatocytes by clathrin-dependent endocytosis.
J. Virol.
86
9443
9453
Hyttinen
JM
,
Niittykoski
M
,
Salminen
A
,
Kaarniranta
K
2013
Maturation of autophagosomes and endosomes: a key role for Rab7.
Biochim. Biophys. Acta
1833
503
510
Inoue
J
,
Ueno
Y
,
Wakui
Y
,
Fukushima
K
,
Kondo
Y
,
Kakazu
E
,
Ninomiya
M
,
Niitsuma
H
,
Shimosegawa
T
2011
Enhanced replication of hepatitis B virus with frameshift in the precore region found in fulminant hepatitis patients.
J. Infect. Dis.
204
1017
1025
Jäger
S
,
Bucci
C
,
Tanida
I
,
Ueno
T
,
Kominami
E
,
Saftig
P
,
Eskelinen
EL
2004
Role for Rab7 in maturation of late autophagic vacuoles.
J. Cell Sci.
117
4837
4848
Jordens
I
,
Fernandez-Borja
M
,
Marsman
M
,
Dusseljee
S
,
Janssen
L
,
Calafat
J
,
Janssen
H
,
Wubbolts
R
,
Neefjes
J
2001
The Rab7 effector protein RILP controls lysosomal transport by inducing the recruitment of dynein-dynactin motors.
Curr. Biol.
11
1680
1685
Kian Chua
P
,
Lin
MH
,
Shih
C
2006
Potent inhibition of human Hepatitis B virus replication by a host factor Vps4.
Virology
354
1
6
Lambert
C
,
Döring
T
,
Prange
R
2007
Hepatitis B virus maturation is sensitive to functional inhibition of ESCRT-III, Vps4, and gamma 2-adaptin.
J. Virol.
81
9050
9060
Lang
T
,
Lo
C
,
Skinner
N
,
Locarnini
S
,
Visvanathan
K
,
Mansell
A
2011
The hepatitis B e antigen (HBeAg) targets and suppresses activation of the toll-like receptor signaling pathway.
J. Hepatol.
55
762
769
Levine
B
2005
Eating oneself and uninvited guests: autophagy-related pathways in cellular defense.
Cell
120
159
162
Li
J
,
Liu
Y
,
Wang
Z
,
Liu
K
,
Wang
Y
,
Liu
J
,
Ding
H
,
Yuan
Z
2011
Subversion of cellular autophagy machinery by hepatitis B virus for viral envelopment.
J. Virol.
85
6319
6333
Liang
TJ
2009
Hepatitis B: the virus and disease.
Hepatology
49 Suppl.
S13
S21
Macovei
A
,
Petrareanu
C
,
Lazar
C
,
Florian
P
,
Branza-Nichita
N
2013
Regulation of hepatitis B virus infection by Rab5, Rab7, and the endolysosomal compartment.
J. Virol.
87
6415
6427
McMahon
BJ
2005
Epidemiology and natural history of hepatitis B. Semin.
Liver Dis.
25 Suppl. 1
3
8
Milich
D
,
Liang
TJ
2003
Exploring the biological basis of hepatitis B e antigen in hepatitis B virus infection.
Hepatology
38
1075
1086
Ozasa
A
,
Tanaka
Y
,
Orito
E
,
Sugiyama
M
,
Kang
JH
,
Hige
S
,
Kuramitsu
T
,
Suzuki
K
,
Tanaka
E
,
Okada
S
et al. 
2006
Influence of genotypes and precore mutations on fulminant or chronic outcome of acute hepatitis B virus infection.
Hepatology
44
326
334
Pankiv
S
,
Alemu
EA
,
Brech
A
,
Bruun
JA
,
Lamark
T
,
Overvatn
A
,
Bjørkøy
G
,
Johansen
T
2010
FYCO1 is a Rab7 effector that binds to LC3 and PI3P to mediate microtubule plus end-directed vesicle transport.
J. Cell Biol.
188
253
269
Prange
R
2012
Host factors involved in hepatitis B virus maturation, assembly, and egress.
Med. Microbiol. Immunol. (Berl.)
201
449
461
Rijckborst
V
,
Sonneveld
MJ
,
Janssen
HL
2011
Review article: chronic hepatitis B – anti-viral or immunomodulatory therapy?
Aliment. Pharmacol. Ther.
33
501
513
Schroeder
B
,
Weller
SG
,
Chen
J
,
Billadeau
D
,
McNiven
MA
2010
A Dyn2-CIN85 complex mediates degradative traffic of the EGFR by regulation of late endosomal budding.
EMBO J.
29
3039
3053
Schroeder
B
,
Srivatsan
S
,
Shaw
A
,
Billadeau
D
,
McNiven
MA
2012
CIN85 phosphorylation is essential for EGFR ubiquitination and sorting into multivesicular bodies.
Mol. Biol. Cell
23
3602
3611
Schulze
RJ
,
Weller
SG
,
Schroeder
B
,
Krueger
EW
,
Chi
S
,
Casey
CA
,
McNiven
MA
2013
Lipid droplet breakdown requires dynamin 2 for vesiculation of autolysosomal tubules in hepatocytes.
J. Cell Biol.
203
315
326
Sir
D
,
Tian
Y
,
Chen
WL
,
Ann
DK
,
Yen
TS
,
Ou
JH
2010
The early autophagic pathway is activated by hepatitis B virus and required for viral DNA replication.
Proc. Natl. Acad. Sci. USA
107
4383
4388
Stenmark
H
2009
Rab GTPases as coordinators of vesicle traffic.
Nat. Rev. Mol. Cell Biol.
10
513
525
Stieler
JT
,
Prange
R
2014
Involvement of ESCRT-II in hepatitis B virus morphogenesis.
PLoS ONE
9
e91279
Tian
Y
,
Sir
D
,
Kuo
CF
,
Ann
DK
,
Ou
JH
2011
Autophagy required for hepatitis B virus replication in transgenic mice.
J. Virol.
85
13453
13456
van der Kant
R
,
Fish
A
,
Janssen
L
,
Janssen
H
,
Krom
S
,
Ho
N
,
Brummelkamp
T
,
Carette
J
,
Rocha
N
,
Neefjes
J
2013
Late endosomal transport and tethering are coupled processes controlled by RILP and the cholesterol sensor ORP1L.
J. Cell Sci.
126
3462
3474
Vanlandingham
PA
,
Ceresa
BP
2009
Rab7 regulates late endocytic trafficking downstream of multivesicular body biogenesis and cargo sequestration.
J. Biol. Chem.
284
12110
12124
Wang
T
,
Ming
Z
,
Xiaochun
W
,
Hong
W
2011
Rab7: role of its protein interaction cascades in endo-lysosomal traffic.
Cell. Signal.
23
516
521
Watanabe
T
,
Sorensen
EM
,
Naito
A
,
Schott
M
,
Kim
S
,
Ahlquist
P
2007
Involvement of host cellular multivesicular body functions in hepatitis B virus budding.
Proc. Natl. Acad. Sci. USA
104
10205
10210
Wieland
SF
,
Chisari
FV
2005
Stealth and cunning: hepatitis B and hepatitis C viruses.
J. Virol.
79
9369
9380
Yan
H
,
Zhong
G
,
Xu
G
,
He
W
,
Jing
Z
,
Gao
Z
,
Huang
Y
,
Qi
Y
,
Peng
B
,
Wang
H
et al. 
2012
Sodium taurocholate cotransporting polypeptide is a functional receptor for human hepatitis B and D virus.
eLife
1
e00049

Competing interests

The authors declare no competing or financial interests.

Supplementary information