Here, we reveal that DOCK11 regulated the GTPase-activating protein Arf-GAP with GTPase, ankyrin repeat, and pleckstrin homology domain-containing protein 2 (AGAP2), which plays important roles in the retrograde trafficking of cargo from EEs to the TGN, subsequently facilitating HBV retrograde transport from the EE-TGN-endoplasmic reticulum (ER) pathway to the nucleus for viral genome replication. A combinatorial treatment regimen incorporating entecavir (ETV) and DOCK11 ablation significantly restricted cccDNA levels in vitro. These observations suggest an unrecognized trafficking route of HBV from the cell surface to the nucleus via the TGN, which may facilitate the maintenance of cccDNA, and highlight DOCK11 as a potential therapeutic target for persistent HBV infection.
Discussion
In this study, we found that HBV used a unique retrograde trafficking route via the EE-TGN-ER pathway to the cell nucleus that had not been implicated previously in HBV infection. This trafficking route could be beneficial for HBV by avoiding lysosomal degradation and thereby facilitating the maintenance of cccDNA in the nucleus. Interestingly, we showed that DOCK11 accelerated this pathway.
DOCK11 is a member of the Zizimin/DOCK family of GEFs and is a prominent GEF for CDC42.
6- Lin Q.
- Yang W.
- Baird D.
- Feng Q.
- Cerione R.A.
Identification of a DOCK180-related guanine nucleotide exchange factor that is capable of mediating a positive feedback activation of Cdc42.
Moreover, the activation of CDC42 together with its downstream target WASP and the ɣCOP subunit of the COPI complex influences F-actin organization and Golgi apparatus integrity and regulates Golgi-to-ER transport.
19- Spang A.
- Shiba Y.
- Randazzo P.A.
Arf GAPs: gatekeepers of vesicle generation.
, 20- Luna A.
- Matas O.B.
- Martinez-Menarguez J.A.
- Mato E.
- Duran J.M.
- Ballesta J.
- Way M.
- Egea G.
Regulation of protein transport from the Golgi complex to the endoplasmic reticulum by CDC42 and N-WASP.
However, a recent report showed that CDC42 appears to promote intra-Golgi anterograde transport rather than affecting COPI transport back to the ER.
21- Park S.Y.
- Yang J.S.
- Schmider A.B.
- Soberman R.J.
- Hsu V.W.
Coordinated regulation of bidirectional COPI transport at the Golgi by CDC42.
Therefore, it is unlikely that CDC42 is involved directly in the retrograde nuclear trafficking of HBV via the EE-TGN-ER pathway identified in this study.
To clarify the direct mechanism by which DOCK11 participates in the retrograde trafficking of HBV, we identified AGAP2 as a novel partner of DOCK11 using liquid chromatography-tandem mass spectrometry. AGAP2 is a crucial regulator of retrograde transport together with ARF1, a representative GTP-binding protein involved in retrograde protein trafficking of STX, cholera toxin, and the endogenous proteins TGN46 and mannose 6-phosphate receptor.
16- Shiba Y.
- Romer W.
- Mardones G.A.
- Burgos P.V.
- Lamaze C.
- Johannes L.
AGAP2 regulates retrograde transport between early endosomes and the TGN.
ARF1 changes between active GTP-bound and inactive GDP-bound conformations, depending on GEFs for activation and GTPase-activating proteins for inactivation.
19- Spang A.
- Shiba Y.
- Randazzo P.A.
Arf GAPs: gatekeepers of vesicle generation.
Notably, we showed that DOCK11 and AGAP2 formed a complex and could bind to ARF1, DOCK11 exhibited GEF activity, and AGAP2 had GAP activity toward ARF1 (
Figures 9 and
10). It was interesting that DOCK11 induced AGAP2 expression, and they formed a complex, which might be beneficial for recruiting ARF1 to this complex. ARF1 was activated by DOCK11 and deactivated by AGAP2 continuously, which would facilitate the efficient recycling of vesicles and cargo release during HBV retrograde trafficking (
Figure 10). Moreover, we showed that DOCK11 could bind to HBcAg, which is also advantageous for DOCK11 to facilitate the recruitment of HBV capsids to retrograde trafficking (
Figure 7). ARF1 regulates COPI vesicles that mediate transport between Golgi stacks or from the Golgi to the ER.
22- Kartberg F.
- Asp L.
- Dejgaard S.Y.
- Smedh M.
- Fernandez-Rodriguez J.
- Nilsson T.
- Presley J.F.
ARFGAP2 and ARFGAP3 are essential for COPI coat assembly on the Golgi membrane of living cells.
We also showed that the depletion of COPI could prevent NL-HBV capsid transport into the nucleus of RAB7KO cells (
Figure 12), indicating the involvement of the trafficking route from the TGN to ER for HBV infection.
Previous reports showed that HBV enters cells via endocytosis and subsequently travels from EEs to lysosomes via RAB7, where uncoating of the virus occurs and HBV degradation is facilitated.
9- Macovei A.
- Petrareanu C.
- Lazar C.
- Florian P.
- Branza-Nichita N.
Regulation of hepatitis B virus infection by Rab5, Rab7, and the endolysosomal compartment.
In this study, we showed that RAB7A was not necessary for HBV infection but rather decreased HBV infection (
Figure 4). RAB7KO cells were more suspectable to HBV infection, as shown by the increased intracellular levels of HBV DNA and cccDNA after infection with HBV particles from HepAD38 cells (
Figure 4D) or NL-HBV particles (
Figure 4E and
F). This finding was also supported by the increased PLA fluorescent punctate intracellular signals in RAB7KO cells (
Figure 5). Thus, we showed that the retrograde trafficking route through the EE-TGN-ER pathway to the nucleus could be more efficient than that previously recognized through the EE-LE pathway to the nucleus.
For many viruses, acidification-mediated proteinase activation in lysosomes is helpful for uncoating and the establishment of infection. However, HBV infection is pH-independent
23- Hagelstein J.
- Fathinejad F.
- Stremmel W.
- Galle P.R.
pH-independent uptake of hepatitis B virus in primary human hepatocytes.
and possibly mediated by translocation motifs,
24- Stoeckl L.
- Funk A.
- Kopitzki A.
- Brandenburg B.
- Oess S.
- Will H.
- Sirma H.
- Hildt E.
Identification of a structural motif crucial for infectivity of hepatitis B viruses.
which may preserve viral morphology. In this study, lysosome inhibitors did not reduce HBV infection, which was compatible with previous findings.
23- Hagelstein J.
- Fathinejad F.
- Stremmel W.
- Galle P.R.
pH-independent uptake of hepatitis B virus in primary human hepatocytes.
,25Duck hepatitis B virus infection of hepatocytes is not dependent on low pH.
Therefore, conformational changes of the viral envelope rather than its extensive degradation might be sufficient for HBV uncoating.
Super-resolution microscopy of fluorescently labeled capsid and surface antigens of infected HBV particles suggested that the uncoating of envelope protein might occur at a relatively early phase after or during trafficking from EEs to LEs or EEs to the TGN (
Figure 2). Interestingly, in duck HBV, TGN host proteins such as carboxypeptidase D
26- Kuroki K.
- Eng F.
- Ishikawa T.
- Turck C.
- Harada F.
- Ganem D.
gp180, a host cell glycoprotein that binds duck hepatitis B virus particles, is encoded by a member of the carboxypeptidase gene family.
and furin
27- Tong Y.
- Tong S.
- Zhao X.
- Wang J.
- Jun J.
- Park J.
- Wands J.
- Li J.
Initiation of duck hepatitis B virus infection requires cleavage by a furin-like protease.
participate in viral entry and cleavage of the large envelope protein for infection. Therefore, in HBV, Golgi host proteins might be involved in infection, and thus, retrograde trafficking of HBV from EEs to the TGN is not peculiar.
What is the role of retrograde trafficking from the EE-TGN-ER pathway to the nucleus in the pathogenesis of HBV infection? This trafficking route separates cargo from the cytosol by a membrane, which could protect HBV from cytosolic innate immune responses that mediate viral degradation. Moreover, naked viral capsids could be the target of protein degradation pathways such as ubiquitination-proteasome and autophagy. It could be speculated that during active replication, a large amount of virus would go to the classic lysosomal pathway, and the virus that “spills over” from lysosomal degradation continues to replicate. During inactive replication, a small amount of virus would use the retrograde pathway through the Golgi, thereby avoiding lysosomal degradation and maintaining replication. Further study is necessary to reveal the regulation of these 2 pathways.
In the liver of CHB patients, we showed that ETV treatment significantly decreased DOCK11 expression together with decreased HBV replication (
Figure 13A–C). The decreased expression of DOCK11 might be reflected in the reduced replication of HBV in hepatocytes, because we found that DOCK11 levels are increased in primary human hepatocytes by HBV infection. Furthermore, the reduction of HBsAg was significantly correlated with that of DOCK11 (
Figure 13D), indicating that cccDNA could be regulated by DOCK11 in the liver of CHB patients. In addition, the combination of knocking down DOCK11 together with ETV treatment further suppressed HBV replication compared with either factor alone. These findings highlighted the substantial potential for developing therapeutics that target DOCK11 in CHB patients. The possible involvement of DOCK11 in HBV replication should be explored further.
Although our study showed that DOCK11 activated the retrograde trafficking of HBV from the EE-TGN-ER pathway to the cell nucleus, it is not known how the nucleocapsid released from the ER enters the nucleus. Although the intrinsic mechanisms are not yet known, there are connections between the ER and nuclear membrane, and a recent report showed that lipid droplets generated in the ER lumen can move into the nucleoplasm of hepatocytes by a precise molecular mechanism.
28- Soltysik K.
- Ohsaki Y.
- Tatematsu T.
- Cheng J.
- Fujimoto T.
Nuclear lipid droplets derive from a lipoprotein precursor and regulate phosphatidylcholine synthesis.
It was also reported that HBV capsid combined with lipid droplets could efficiently establish infection.
29Lipid-mediated introduction of hepatitis B virus capsids into nonsusceptible cells allows highly efficient replication and facilitates the study of early infection events.
Therefore, ER stress-induced lipid droplets in the ER potentially participate in the retrograde trafficking of HBV via the EE-TGN-ER pathway to the cell nucleus.
DOCK11 may also potentially increase HBV infection by increasing the uptake of HBV at the entry level because endocytosis is mediated by actin polymerization on the cell surface, and this process is regulated by CDC42. Moreover, further studies should be performed to uncover the roles of DOCK11 in cccDNA formation in the nucleus because confocal microscopy revealed the low-level expression of DOCK11 in the nucleus.
30- Ide M.
- Tabata N.
- Yonemura Y.
- Shirasaki T.
- Murai K.
- Wang Y.
- Ishida A.
- Okada H.
- Honda M.
- Kaneko S.
- Doi N.
- Ito S.
- Yanagawa H.
Guanine nucleotide exchange factor DOCK11-binding peptide fused with a single chain antibody inhibits hepatitis B virus infection and replication.
In conclusion, our findings might lead to the identification of new areas in the early steps of the HBV life cycle as well as facilitate the development of molecular targeting therapies to cure HBV infection.
Materials and Methods
Key resources are shown in
Table 1, and the oligonucleotides for shRNA, CRISPR/Cas9, cloning, and RTD-PCR are shown in
Table 2.
Table 2Oligonucleotides for shRNA, CRISPR/Cas9, Cloning, and RTD-PCR
Patients
DOCK11 expression was evaluated using non-tumor liver biopsied specimens from 17 HBV-infected HCC patients before and after ETV therapy. HBsAg and HBV DNA levels were evaluated in serum samples isolated by centrifugation of coagulated whole blood and preserved at −80°C. The research protocols were reviewed by the ethics committee at Kanazawa University and its related hospitals. Informed consent was obtained from all patients.
Cell Lines
The human liver cancer Huh7, HepG2-NTCP-C4, HepG2.2.15, and HepAD38 cell lines were authenticated by DNA fingerprinting in 2016. Primary human hepatocytes (PXB cells) and their specific medium were purchased from PhoenixBio Co, Ltd (Hiroshima, Japan). The Huh7 and HepG2.2.15 cell lines were maintained in Dulbecco modified Eagle medium (Life Technologies, Carlsbad, CA), and the other cell lines we established were cultured in Dulbecco modified Eagle medium/F-12, GlutaMAX supplement (Thermo Fisher Scientific, Waltham, MA); both media contained 10% fetal bovine serum, 100 U/mL penicillin, and 100 μg/mL streptomycin. The addition of 400 μg/mL G418, 10 mmol/L HEPES buffer solution, and 5 μg/mL insulin was needed for the culture of the HepG2-NTCP-C4 and HepAD38 cell lines.
HBV Preparation
HepAD38 cell culture supernatant filtered through 0.45-μm VWR filter units was mixed with PEG 8000 (40% wt/vol; Sigma-Aldrich, St Louis, MO) containing 800 mmol/L NaCl to a final concentration of 10% (wt/vol) PEG 8000 and 200 mmol/L NaCl. The mixture was incubated overnight at 4°C, followed by centrifugation at 3500
g for 30 minutes. Pellets were suspended in Opti-MEM reduced serum medium (Invitrogen) to 1/100 of the original volume, and HBV DNA was extracted using an SMI TEST EX R&D Kit (MLB, Nagoya, Japan). HBV copies were detected by RTD-PCR. For the preparation of NL-HBV particles, the pUC1.2xHBV/NL and pUCxHBV-delta plasmids were provided by Professor Shimotohno (Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Chiba, Japan) and co-transfected into HepG2 cells using Lipofectamine 3000 (Invitrogen). Cell culture supernatant was harvested at 5 and 7 days after transfection and filtered through 0.45-μm filter units. Harvested NL-HBV particles were treated by DNase I and used for infection. Viral particles were extracted from the supernatants of HepG2.2.15-HBV-CAT-TC155 cells or HepG2.2.15-HBV-CAT-TCPreS1 cells using a PEG Virus Precipitation Kit and labeled with a TC-ReAsH II In-Cell Tetracysteine Tag Detection Kit (Thermo Fisher Scientific). ReAsH-TC155HBV was tagged with tetracysteine (C-C-P-G-C-C) at the 155th capsid amino acid and labeled with the ReAsH fluorescent dye
15- Sun S.
- Yan J.
- Xia C.
- Lin Y.
- Jiang X.
- Liu H.
- Ren H.
- Yan J.
- Lin J.
- He X.
Visualizing hepatitis B virus with biarsenical labelling in living cells.
; ReAsH-PreS1HBV virions tagged with tetracysteine in PreS1 were labeled with the ReAsH fluorescent dye. The HBV used to infect PXB cells was purchased from PhoenixBio Co, Ltd.
Establishment of Huh7-shDOCK11, HepG2-NTCP-C4-shDOCK11, and HepG2-NTCP-C4-RAB7KO Cell Lines
To establish the Huh7-shDOCK11 and HepG2-NTCP-C4-shDOCK11 cell lines, MISSION Lentiviral Packaging Mix (Sigma-Aldrich) was used to generate lentiviruses with an shRNA plasmid targeting DOCK11 in the Lenti-X 293T Cell Line (Takara Bio USA, Mountain View, CA). As a negative control, the pLKO.1 Puro Non-Target shRNA Control Plasmid was used. Huh7 and HepG2-NTCP-C4 cells were infected with medium containing the lentiviruses generated from Lenti-X 293T cells. At 20 hours after infection, the medium was replaced with fresh medium. At 3 days after infection, the cells were selected by 5 mg/mL puromycin (Thermo Fisher Scientific). All experiments were performed within 2 weeks after lentiviral transduction. To establish the HepG2-NTCP-C4 RAB7KO cell line, the target sequence of RAB7A was cloned into the lentiCRISPRv2 backbone (Addgene, Cambridge, MA). To make the lentiviruses, the plasmid (to clone the target sequences into the lentiCRISPRv2 backbone) was co-transfected into Lenti-X 293T cells (Takara Bio USA) with Packaging Mix (third generation mix from ABM). Harvested lentiviruses were transduced into HepG2-NTCP-C4 cells. At 3 days after transduction, these cells were treated with 5 mg/mL puromycin for 1 week. After puromycin selection, gene knockout of RAB7 was confirmed by immunoblotting and immunofluorescence analyses.
Establishment of the Dox-Inducible HepG2-NTCP-C4-Halo-DOCK11 Cell Line
The fragment of Halo-DOCK11 was digested from the pFN21A Halo-DOCK11 CMV Flexi vector (FHC29467) and inserted into the Retro-X Tet-On 3G vector, followed by the establishment of HepG2-NTCP-C4-Halo-DOCK11 stable cells according to the instructions of the Retro-X Tet-On 3G inducible expression system (Takara Bio USA).
Infection Model of HepG2-NTCP-C4–Derived Cells
HepG2-NTCP-C4-RAB7KO and control HepG2-NTCP-C4-ScrambleKO cells, HepG2-NTCP-C4-shDOCK11 and control HepG2-NTCP-C4-shScramble cells, and HepG2-NTCP-C4-Halo-DOCK11 cells were established from HepG2-NTCP-C4 cells as described below. These cells were infected with recombinant NL-HBV particles or HBV virions from HepAD38 cells at 6,000,000 genome equivalents (GEq)/cell in the presence of 4% PEG 8000 and 2% DMSO. For HBV DNA and cccDNA detection, the cells were infected with NL-HBV virions for 30 hours. For the PLA or immunofluorescence analysis, the cells were infected with NL-HBV virions, ReAsH-TC155HBV, or ReAsH-PreS1HBV virions for 30, 60, 90, 120, and 180 minutes. For the HBcAg enzyme-linked immunosorbent assay and co-immunoprecipitation assay, the cells were infected with HBV virions from HepAD38 cells for 2 hours. In some experiments, at 1 day before infection with HBV virions, the cells were transfected with plasmid or siRNA or treated with Dox.
Infection Model of PXB Cells
As a comparison with HCC cells, freshly isolated hepatocytes from humanized chimeric mouse liver (PXB cells) were used to confirm the function of DOCK11. PXB cells in a special culture medium (#PPC-M200; PhoenixBio Co, Ltd) were infected with HBV particles (#PPC-BC; PhoenixBio Co, Ltd) at 5 GEq/cell in the presence of 4% PEG 8000 and 2% DMSO overnight. The following day, the medium was replaced with fresh medium and then changed every 4 days until day 24. At day 24, total RNA was collected, and DOCK11 mRNA levels were detected by RTD-PCR. In some experiments, PXB cells were infected with lentiviruses carrying shDOCK11, shAGAP2, or control Scramble shRNA. The following day, the medium was replaced with fresh medium. After 7 days, PXB cells were infected with HBV particles (PhoenixBio Co, Ltd) or HBV particles from HepAD38 cells overnight, and the medium was changed every 4 days until day 17. Total RNA and genomic DNA were collected. DOCK11, AGAP2, HBV DNA, and cccDNA were detected by RTD-PCR analysis or Southern blotting. In another experiment, original PXB cells were reseeded into 8-chamber slides. After 7 days, the cells were infected with HBV particles (PhoenixBio Co, Ltd) overnight, and fresh medium was added every 4 days until day 30. Immunofluorescence analysis was carried out using Alexa Fluor 488-labeled DOCK11 together with anti-TGN46, anti-GM130, or anti-HBcAg antibodies as the primary antibody. The secondary antibody was Alexa Fluor 594-conjugated anti-mouse immunoglobulin (Ig) G antibody. Images were captured with a Dragonfly high-speed confocal microscope (Andor, Oxford Instruments, Belfast, UK).
Hirt DNA Extraction and Southern Blot Analysis
The Hirt protein-free DNA extraction procedure was used to isolate cccDNA from HBV-infected cells or HepAD38 cells.
31- Cai D.
- Nie H.
- Yan R.
- Guo J.T.
- Block T.M.
- Guo H.
A southern blot assay for detection of hepatitis B virus covalently closed circular DNA from cell cultures.
Full-length HBV was inserted into a pSPT19 vector to generate the pSPT19-HBV plasmid. The pSPT19-HBV template was linearized by
BamHI (Takara, Shiga, Japan), and in vitro transcription was performed with 1 μg linearized DNA template and a DIG RNA Labeling Kit (Roche, Basel, Switzerland) in the presence of T7 RNA polymerase to generate digoxigenin-UTP-labeled, single-stranded RNA probes. Hirt-extracted DNA was electrophoresed on 1.2% agarose gels and blotted onto a Hybond-N+ membrane (GE Healthcare, Amersham, UK). DIG-labeled single-stranded RNA probes transcribed from pSPT19-HBV plasmids were used to detect HBV cccDNA. Hybridization was performed with a 30-minute pre-hybridization at 50°C in 10 mL DIG Easy Hyb buffer (Roche) and with overnight hybridization at 50°C in 5 mL pre-warmed DIG Easy Hyb buffer containing 1 μg DIG-labeled probe (Roche). The membranes were washed with a Wash and Block Buffer Set (Roche). Probe-target hybrids were localized with 4 μL of anti-digoxigenin-AP conjugate (Roche) and detected by 1 mL CSPD (Roche). Images were acquired with the ChemiDoc Touch Imaging System (Bio-Rad, Hercules, CA).
Iodixanol Density Gradient Analysis
The medium from HepG2-HBV-CAT-TCPreS1 cells was harvested at 6, 8, and 10 days after plating. The virus fraction was precipitated using a PEG Virus Precipitation Kit (BioVision, Milpitas, CA). After determining HBV DNA levels by RTD-PCR, 2.8 × 109 copies of HBV DNA were layered on a 10%–40% iodixanol gradient and centrifuged at 178,000g for 21 hours at 4ºC in an SW41Ti rotor. Fractions were collected from the top of the gradient, and the density and HBsAg, HBcrAg, and HBV DNA levels were measured in each fraction. HepG2-NTCP-C4 cells were infected with fractions 17 and 18, and fluorescence microscopy images were captured with an HM-1000 super-resolution microscope (Sysmex Corp, Kobe, Japan) and Dragonfly confocal microscope (Andor, Oxford Instruments).
Super-Resolution Fluorescence Microscopy
HepG2-NTCP-C4 cells were seeded at a density of 1.0 × 104 cells/mL in Nunc Lab-Tek chamber slides (Capitol Scientific, Austin, TX) and incubated with a baculovirus infection system encoding GFP-fused RAB5A, GFP-fused RAB7A, and CellLight Golgi-GFP BacMam 2.0 (Thermo Fisher Scientific) overnight at 37°C in a 5% CO2 atmosphere. The next day, the medium was replaced with fresh medium containing Hoechst 33342 (Thermo Fisher Scientific) for another 30 minutes. The medium was replaced by fresh growth medium containing ReAsH-labeled TC155HBV particles or ReAsH-labeled TCPreS1HBV particles purified by iodixanol density gradient. At the indicated times, the cells were fixed in 4% paraformaldehyde. Images were captured with an HM-1000 super-resolution microscope (Sysmex Corp) and Dragonfly confocal microscope (Andor, Oxford Instruments).
Live Cell Confocal Imaging
The cells were seeded at a density of 1.0 × 104 cells/mL in chamber slides and incubated with CellLight Golgi-GFP BacMam 2.0 or CellLight ER-GFP BacMam 2.0 overnight at 37°C in a 5% CO2 atmosphere. The next day, the medium was replaced with fresh growth medium containing Hoechst 33342 for another 30 minutes. Then, the medium was replaced with fresh growth medium containing ReAsH-labeled TC155HBV particles. Time-lapse fluorescence images of ReAsH-labeled TC155HBV particles were acquired every 1 minute for up to 180 minutes (180 frames) using an LSM510 inverted confocal microscope (Zeiss, Jena, Germany).
Constructs and Transfection of Plasmids
The Halo-DOCK11 (FHC29467) and Halo-AGAP2 (FHC24787) plasmids were purchased from the Kazusa DNA Research Institute (Chiba, Japan). The plasmid constructs for AGAP2-Flag and DHR2-DOCK11-Flag were cloned into pcDNA3.1 using the primer sets shown in
Table 2. Transfections were performed with Lipofectamine 3000 (Invitrogen).
Dynasore, Pitstop 2, Retro-2, BFA, and NAV-2729 Treatment of HepG2-NTCP-C4 or RAB7KO Cells Infected With Recombinant NL-HBV Particles
HepG2-NTCP-C4 cells transfected with Halo-DOCK11 or HepG2-NTCP-C4 RAB7KO cells were infected with recombinant NL-HBV particles or HBV from HepAD38 cells at 6,000,000 GEq/cell with or without Dynasore, Pitstop 2, Retro-2, BFA, or NAV-2729 at the indicated concentrations. All infections were performed in the presence of 4% PEG 8000 and 2% DMSO at 37°C for 30 hours. After the cells were washed with phosphate-buffered saline (PBS), cell lysate and genomic DNA were collected. HBV DNA and cccDNA levels were determined by RTD-PCR, and Luc fluorescence was detected by a Nano-Glo Luciferase Assay Kit (Promega, Madison, WI).
Lysosome Inhibitor Treatment of HepG2-NTCP-C4 Cells Infected With Recombinant NL-HBV Particles
HepG2-NTCP-C4 cells were infected with recombinant NL-HBV particles at 60,000 GEq/cell with or without lysosome inhibitors (Sigma-Aldrich) including E46d, Baf1, chloroquine, or NH4Cl at the indicated concentrations. All infections were performed in the presence of 4% PEG 8000 and 2% DMSO at 37°C for 30 hours. After the cells were washed with PBS, the cell lysate and genomic DNA were collected. HBV DNA levels were determined by RTD-PCR. Cell density was evaluated using a Cell Counting Kit-8 (Dojindo, Kumamoto, Japan).
Establishment of the HepAD38-shDOCK11 and HepG2.2.15-shDOCK11 Cell Lines
MISSION Lentiviral Packaging Mix (Sigma-Aldrich) was used to generate lentiviruses with an shRNA plasmid targeting DOCK11 in Lenti-X 293T cells (Takara Bio USA). As a negative control, the pLKO.1 Puro Non-Target shRNA Control Plasmid was used. HepAD38 and HepG2.2.15 cells were infected with medium containing the lentiviruses generated from Lenti-X 293T cells. At 24 hours after infection, the medium was replaced with fresh medium. At 3 days after infection, the cells were selected by 8 mg/mL puromycin (Thermo Fisher Scientific).
Immunofluorescence Analysis
The cells were seeded at 1.0 × 105 cells/well in a 4-chamber slide, and after 16–18 hours, they were transfected with the GFP-DOCK11 plasmid or treated with Dox for HepG2-NTCP-C4-Halo-DOCK11 cells. At 48 hours after transfection, the cells were washed 3 times with PBS and fixed in 4% paraformaldehyde for 10 minutes at room temperature. After washing with PBS-T, the cells were permeabilized with PBS containing 0.5% Triton X-100, incubated in a blocking solution (X0909; Dako, Santa Clara, CA) for 30 minutes, and reacted with an anti-GFP antibody (cat. no. 598; Medical and Biological Lab Co, Ltd, Aichi, Japan) and Alexa Fluor 555-labeled DOCK11 as the primary antibody for 1 hour. The secondary antibody was Alexa Fluor 488-conjugated anti-rabbit Ig G antibody for GFP. The cells were mounted with mounting medium containing DAPI (Vector Laboratories, Burlingame, CA). Imaging was performed with a CSU-X1 confocal microscope (Yokogawa Electric Corporation, Tokyo, Japan). In some experiments, HepG2-NTCP-C4 RAB7KO cells were seeded at 2.0 × 104 cells/well in an 8-chamber slide, and at 16–18 hours later, the cells were infected with NL-HBV particles. At the indicated times, the cells were fixed in 4% paraformaldehyde. Images were captured with a Dragonfly high-speed confocal microscope (Andor, Oxford Instruments).
PLA
To visualize the close proximity of HBcAg and EEs, TGN, or ER, HepG2-NTCP-C4 or HepG2-NTCP-C4-RAB7KO cells were infected with NL-HBV particles at a multiplicity of infection of 6,000,000 and incubated for the indicated times before fixation with 4% paraformaldehyde. After fixation, the cells were permeabilized with PBS containing 0.5% Triton X-100 and then incubated in a blocking solution (X0909; Dako) for 30 minutes. The cells were stained for HBcAg (1:100 dilution), RAB5 (1:100 dilution), TGN46 (1:50 dilution), or PDI (1:100 dilution) by overnight incubation at 4°C. These cells were immunostained using the PLA (Sigma-Aldrich). The cells were washed, and the nuclei were counterstained with DAPI. Fluorescence micrographs were collected by confocal microscopy. Fluorescent foci were quantified using ImageJ software (available from
http://rsb.info.nih.gov/ij/). Threshold levels were set using a PLA-processed sample without primary antibodies in each staining group.
Extraction of the Golgi Fraction
The Golgi fraction was extracted from cultured cells using a Golgi Isolation Kit (Sigma-Aldrich). The Golgi fraction was verified by immunoblotting with a rabbit anti-human GS28 antibody. For the HBcAg enzyme-linked immunosorbent assay, HepG2-NTCP-C4 RAB7KO cells were treated with BFA at 30 minutes before infection and then replacement with fresh medium containing NL-HBV virions, 4% PEG 8000, 2% DMSO, and 2 μmol/L BFA. After infection for 2 hours and BFA treatment, the cells were harvested, and the Golgi fraction was extracted.
HBcAg Enzyme-Linked Immunosorbent Assay
The HepG2-NTCP-C4-ScrambleKO and HepG2-NTCP-C4-RAB7KO cell lines were infected with HBV particles purified from the HepAD38 cell line. After infection for 3 hours, the Golgi fraction was isolated (GL0010; Sigma-Aldrich). HBcAg protein levels were measured by a commercial enzyme-linked immunosorbent assay kit (ARG82006; Arigo, Taiwan, China).
Immunoblotting Analysis
Immunoblotting analysis was performed as described previously.
32- Murai K.
- Honda M.
- Shirasaki T.
- Shimakami T.
- Omura H.
- Misu H.
- Kita Y.
- Takeshita Y.
- Ishii K.A.
- Takamura T.
- Urabe T.
- Shimizu R.
- Okada H.
- Yamashita T.
- Sakai Y.
- Kaneko S.
Induction of selenoprotein P mRNA during hepatitis C virus infection inhibits RIG-I-mediated antiviral immunity.
Protein extracts were prepared with RIPA Lysis Buffer (Merck Millipore, Burlington, MA) containing Protease Inhibitor Cocktail and Phosphatase Inhibitor Tablets (Roche Applied Science, Pleasanton, CA). Halo-DOCK11, RAB7, RAB5, RAB9, RAB11, AGAP2-Flag, ARF1, and β-actin expression was evaluated with mouse anti-Halo (Promega), rabbit anti-RAB7, -RAB5, -RAB9, and -RAB11, rabbit anti-Flag, rabbit anti-ARF1, and rabbit anti-β-actin antibodies (Cell Signaling Technology, Danvers, MA), respectively.
Immunoprecipitation Assay
Cell lysates were incubated with rabbit IgG or rabbit anti-DOCK11, anti-ARF1, or anti-Flag antibodies overnight at 4°C. Immunoprecipitation was performed using Protein G Mag Sepharose (GE Healthcare, Little Chalfont, UK). The beads were washed in lysis buffer and eluted in sodium dodecyl sulfate sample buffer. The reaction mixtures were analyzed by immunoblotting.
Quantification of HBV DNA and cccDNA by RTD-PCR
HBV DNA was extracted from the cells using a DNeasy Blood & Tissue Kit (QIAGEN, Valencia, CA). The extracted DNA (250 ng) was treated for 30 minutes at 37°C with 25 U T5 Exonuclease (New England BioLabs, Ipswich, MA) and treated for 5 minutes at 95°C for enzyme heat inactivation. HBV DNA and cccDNA levels were quantified with qPCR MasterMix Plus Low ROX (Nippon Gene, Tokyo, Japan) using a specific HBV DNA probe (5′-FAM/TATCGCTGG/ZEN/ATGTGTCTGCGGCGT/3IBFQ-3′) and cccDNA probe (5′-FAM-CTGTAGGCATAAATTGGT-MGB-3′). HBV DNA and cccDNA primer sets are shown in
Table 2.
RNA Extraction and RTD-PCR Analysis
Total RNA was isolated using a NIPPON Cell & Tissue RNA Kit (Nippon Gene). A High-Capacity cDNA Reverse Transcription Kit (Thermo Fisher Scientific) was used for first-strand cDNA synthesis with 0.2 μg total RNA from each sample. RTD-PCR was conducted using TaqMan Gene Expression Assay Identification. Quantitative gene expression data were normalized to the expression levels of the housekeeping gene GAPDH (Thermo Fisher Scientific).
Recombinant Proteins
Recombinant DOCK11-DHR2-Flag (containing residues 4678–6285 of DOCK11) was provided by Professor Matsushima (Tokyo University); ARF1, AGAP2, and CDC42 proteins were obtained from Abnova (Taiwan, China).
GEF and GAP Activity Assays in Vitro
GEF and GAP activity assays were performed using a GTPase-Glo Assay Kit (Promega). Briefly, for in vitro GEF activity assays, we prepared a 2× GTPase-GAP solution containing 1 mmol/L DTT and a fixed amount of ARF1 or CDC42 and serially diluted DOCK11 as a GEFase at twice the desired final concentration in GEF buffer (50 mmol/L Tris-HCl, pH 7.5, 50 mmol/L NaCl, 1 mmol/L EDTA, and 10 mmol/L MgCl2). For in vitro GAP activity assays, we prepared a 2× GTPase solution containing 1 mmol/L DTT and a fixed amount of ARF1 and serially diluted AGAP2 as a GAP at twice the desired final concentration in GTPase/GAP buffer (50 mmol/L Tris-HCl, pH 7.5, 50 mmol/L NaCl, 20 mmol/L EDTA, and 5 mmol/L MgCl2). Both reaction solutions were added to an equal volume of GTPase-Glo reagent containing an enzyme to convert the GTP remaining after the GTPase reaction to ATP. The ATP generated in the reaction was detected by a luciferase/luciferin-based reagent. GAP and GEF activity was inversely correlated to the amount of light produced.
Statistical Analysis
All data are presented as the mean ± standard deviation. Experiments were repeated at least 3 times. Two-tailed unpaired Student t test or one-way analysis of variance was used to evaluate the data. Pearson’s and Manders’ correlation coefficients were used to assess colocalization. Spearman’s correlation analysis was used to determine the correlation of the reduction rate of HBsAg and DOCK11. GraphPad Prism 7 software (version 17.0; IBM Corp., Armonk, NY) was used for data analysis. Statistical differences were considered significant at P < .05.
Acknowledgments
The authors thank Professor Yasuhito Tanaka, Department of Gastroenterology, Kumamoto University, for providing the HBV/C vectors and Dr Koichi Watashi, NIH, Japan, for providing the HepG2-NTCP-C4 cells.
CRediT Authorship Contributions
Ying-Yi Li (Data curation: Lead)
Kazuyuki Kuroki (Data curation: Supporting)
Tetsuro Shimakami (Data curation: Supporting)
Kazuhisa Murai (Data curation: Supporting)
Kazunori Kawaguchi (Data curation: Supporting)
Takayoshi Shirasaki (Investigation: Supporting)
Kouki Nio (Data curation: Supporting)
Saiho Sugimoto (Data curation: Supporting)
Tomoki Nishikawa (Data curation: Supporting)
Hikari Okada (Data curation: Supporting)
Noriaki Orita (Data curation: Supporting)
Hideo Takayama (Data curation: Supporting)
Ying Wang (Data curation: Supporting)
Phuong Doan Thi Bich (Data curation: Supporting)
Astuya Ishida (Data curation: Supporting)
Sadahiro Iwabuchi (Methodology: Supporting)
Shinichi Hashimoto (Investigation: Equal)
Takeshi Shimaoka (Resources: Equal)
Noriko Tabata (Methodology: Equal)
Miho Watanabe-Takahashi (Resources: Supporting)
Kiyotaka Nishikawa (Resources: Supporting)
Hiroshi Yanagawa (Data curation: Supporting)
Motoharu Seiki (Conceptualization: Supporting)
Kouji Matsushima (Conceptualization: Supporting)
Taro Yamashita (Data curation: Supporting)
Shuichi Kaneko (Conceptualization: Supporting)
Masao Honda, PhD, MD (Conceptualization: Lead; Writing – original draft: Equal)
Article info
Publication history
Published online: October 18, 2022
Accepted:
October 12,
2022
Received:
December 24,
2021
Footnotes
Conflicts of interest The authors disclose no conflicts.
Funding Supported by the Japan Agency for Medical Research and Development under grant numbers JP20fk0210073, JP20fk0210048, JP20fk0210034, JP20fk0210046, JP20fk0210081, JP20fk0310110, JP20fk0210049, and 18K07966.
Copyright
© 2022 The Authors. Published by Elsevier Inc. on behalf of the AGA Institute.