Coronavirus cell entry occurs through the endo-/lysosomal pathway in a proteolysis-dependent manner

Christine Burkard, Monique H Verheije, Oliver Wicht, Sander I van Kasteren, Frank J van Kuppeveld, Bart L Haagmans, Lucas Pelkmans, Peter J M Rottier, Berend Jan Bosch, Cornelis A M de Haan, Christine Burkard, Monique H Verheije, Oliver Wicht, Sander I van Kasteren, Frank J van Kuppeveld, Bart L Haagmans, Lucas Pelkmans, Peter J M Rottier, Berend Jan Bosch, Cornelis A M de Haan

Abstract

Enveloped viruses need to fuse with a host cell membrane in order to deliver their genome into the host cell. While some viruses fuse with the plasma membrane, many viruses are endocytosed prior to fusion. Specific cues in the endosomal microenvironment induce conformational changes in the viral fusion proteins leading to viral and host membrane fusion. In the present study we investigated the entry of coronaviruses (CoVs). Using siRNA gene silencing, we found that proteins known to be important for late endosomal maturation and endosome-lysosome fusion profoundly promote infection of cells with mouse hepatitis coronavirus (MHV). Using recombinant MHVs expressing reporter genes as well as a novel, replication-independent fusion assay we confirmed the importance of clathrin-mediated endocytosis and demonstrated that trafficking of MHV to lysosomes is required for fusion and productive entry to occur. Nevertheless, MHV was shown to be less sensitive to perturbation of endosomal pH than vesicular stomatitis virus and influenza A virus, which fuse in early and late endosomes, respectively. Our results indicate that entry of MHV depends on proteolytic processing of its fusion protein S by lysosomal proteases. Fusion of MHV was severely inhibited by a pan-lysosomal protease inhibitor, while trafficking of MHV to lysosomes and processing by lysosomal proteases was no longer required when a furin cleavage site was introduced in the S protein immediately upstream of the fusion peptide. Also entry of feline CoV was shown to depend on trafficking to lysosomes and processing by lysosomal proteases. In contrast, MERS-CoV, which contains a minimal furin cleavage site just upstream of the fusion peptide, was negatively affected by inhibition of furin, but not of lysosomal proteases. We conclude that a proteolytic cleavage site in the CoV S protein directly upstream of the fusion peptide is an essential determinant of the intracellular site of fusion.

Conflict of interest statement

The authors have declared that no competing interests exist.

Figures

Figure 1. RNAi-mediated downregulation of endocytosis-associated proteins…
Figure 1. RNAi-mediated downregulation of endocytosis-associated proteins affects MHV infection.
A) Confirmation of endocytosis-associated hits from druggable genome-wide siRNA screen. Gene silencing was performed using individual transfection of three different siRNAs per gene in HeLa-mCC1a cells. Cells were infected with MHV-EGFPM at MOI = 0.5 for 8 h and analyzed by FACS for cell viability and virus replication. The effect of downregulation of expression on MHV infection was studied for the actin cytoskeleton-associated proteins ACTR2 and ACTR3 (orange), late endosomal proteins RAB7A and RAB7B (turquoise), HOPS complex sububit VPS39 (light green), ER/Golgi secretion-associated protein SNX1, Integrin/Actin-associated protein VCL, and Serine/Threonine-protein kinase PAK1 (grey). Error bars represent SEM, n = 4. B) Confirmation of siRNA-mediated reduction in mRNA levels. mRNA levels at 72 h post transfection were measured by qRT-PCR in comparison to non-transfected cells. Error bars represent SEM, n = 3*3. C) The effect of the RNAi-mediated downregulation of an extended set of endocytosis-associated proteins on MHV infection. Infection of MHV-EGFPM was analyzed after downregulation of proteins associated with caveolae-mediated endocytosis (light blue), clathrin-mediated endocytosis (dark blue), early endosomes (cerulean), actin cytoskeleton (dark orange), microtubule cytoskeleton (orange), late endosomes (turquoise), and late endosome-to-lysosome trafficking (light green) as described above. Error bars represent SEM, n = 3. A, C) Dotted lines show the lower 95% confidence interval of the negative siRNA controls.
Figure 2. Endocytosis affecting agents indicate clathrin-mediated…
Figure 2. Endocytosis affecting agents indicate clathrin-mediated endocytosis and endosome maturation to be important in MHV infection.
HeLa-mCC1a cells, inoculated with MHV-EGFPM at MOI = 0.5, were treated with the different inhibitors from 30 min prior to 8 h post inoculation (0–8 h) or from 2–8 h post inoculation (2–8 h; hatched bars): ammonium chloride (NH4Cl), Bafilomycin A1 (BafA1), Chloroquine (Chloq), Chlorpromazine (Chlopro), Monensin (Mon), Dynasore, Dyngo-4A, EIPA, Latrunculin A (LatA), Jasplakinolide (Jasp), Cytochalasin B (CytoB), Cytochalasin D (DytoD), Nocodazole (Noc), MG132, Brefeldin A (BrefA), as well as solvents dimethyl sulfoxide (DMSO) and methanol (MeOH). Infection was determined by FACS and displayed relative to the infection level observed in mock-treated cells (UNTR). Error bars represent SEM, n = 3.
Figure 3. Clathrin-mediated endocytosis and late endosome-to-lysosome…
Figure 3. Clathrin-mediated endocytosis and late endosome-to-lysosome trafficking is required for MHV fusion.
A) Fusion assay upon siRNA-mediated gene silencing. Three different siRNAs per gene were transfected individually into HeLa-mCC1a-ΔM15. 72 h post transfection, cells were pre-loaded with FDG by hypotonic shock. MHV-αN was allowed to bind to the cells on ice at MOI = 20 for 90 min. 100 min post warming to 37°C, cells were collected and analyzed by FACS. Fusion was determined relative to the number of FIC-positive cells observed upon mock treatment of infected cells (UNTR). Error bars represent SEM, n = 3. B) Fusion of MHV upon treatment of cells with different inhibitors was studied as in A. Cells were pretreated with ammonium chloride (NH4Cl), Bafilomycin A1 (BafA1), Chloroquine (Chloq), Chlorpromazine (Chlopro), Monensin (Mon), Dynasore, Dyngo-4A, EIPA, Latrunculin A, (LatA), Jasplakinolide (Jasp), Cytochalasin B (CytoB), Cytochalasin D (DytoD), Nocodazole (Noc), U18666A, MG132, Brefelding A (BrefA), as well as with the solvents dimethyl sulfoxide (DMSO) and methanol (MeOH), protein synthesis inhibitor cyclohexamide (CHX), and MHV fusion inhibitor HR2 peptide (HR2) for 30 min at 37°C. The inhibitors were kept present during binding of MHV-αN to cells and during warming to 37°C cells for 100 min. Fusion was determined relative to the number of FIC-positive cells after mock treatment (UNTR). Error bars represent SEM, n = 3.
Figure 4. Live-cell microscopy demonstrates co-localization and…
Figure 4. Live-cell microscopy demonstrates co-localization and co-tracking of MHV with endosomal vesicles and fusion of MHV in these vesicles.
HeLa-mCC1a cells transfected with plasmids encoding RAB5-mRFP, RAB7-mRFP, or dsRed-LAMP1 were inoculated with DyLight 488-labeled MHV. Live cell imaging was performed to track internalized particles. A) Examples of MHV particles co-localizing with RAB5-, RAB7-, and LAMP1-positive endosomal vesicles. Size bars indicate 0.2 µM B) Virus particles that could be tracked were classified as ‘fusing’ (Fusing) ‘associating/dissociating’ (Assoc/Dissoc), or ‘non-fusing’ (Non-fusing) as described in the Materials and Methods section.
Figure 5. MHV infection depends on endosomal…
Figure 5. MHV infection depends on endosomal maturation.
A) HeLa-mCC1a cells were pretreated with increasing concentrations of Bafilomycin A1 (BafA1) for 30 min and subsequently infected with luciferase expressing MHV, VSV, or IAV in the presence of BafA1. Infection levels were determined by assaying the luciferase activity in cell lysates relative to lysates of infected cells that had been mock treated. Error bars represent SEM, n = 3*3. B) Haploid cells (HAP1), haploid cells lacking VPS33A (H1-ΔV33) or VPS33A-lacking haploid cells retransfected with FLAG-tagged VLP33A (H1-ΔV33-fV33) were infected with luciferase expressing MHV, VSV, or IAV. Cells were lysed at 7 h (MHV and VSV) or 16 h post infection. Infection is displayed relative to virus-driven luciferase expression levels in HAP1 cells. Error bars represent SEM, n = 3*3.
Figure 6. Inhibition of lysosomal proteases prevents…
Figure 6. Inhibition of lysosomal proteases prevents MHV fusion.
The MHV fusion assay was performed on HeLa-mCC1a-ΔM15 cells as described in the legend to Figure 3, in the presence of the protease inhibitors CPI, AEBSF, Aprotinin, Leupeptin, Pepstatin A, Camostat, and Phosphoramidon. As controls, cells were treated with solvent DMSO, MHV fusion inhibitor HR2 peptide (HR2), and lysosomotropic agent ammonium chloride (NH4Cl). Fusion was determined relative to the number of FIC-positive cells after mock treatment (UNTR). Error bars represent SEM, n = 3.
Figure 7. Introduction of a furin cleavage…
Figure 7. Introduction of a furin cleavage site just upstream of the fusion peptide renders MHV independent of lysosomal proteases.
A) Schematic representation of the MHV spike protein. The MHV S proteins are partially processed by furin at the S1/S2 boundary (S1/S2) as indicated by the arrow. The furin cleavage site sequence at this position (RRAHR) is shown. The signal sequence (SS) at the amino-terminal end of the S1 subunit and the approximate positions of the fusion peptide (FP), heptad repeat regions 1 and 2 (HR1 and HR2) and the transmembrane domain (TM) in the S2 subunit are indicated. MHV-S2′FCS virus contains an optimal furin cleavage site (RRRRR) immediately upstream of the FP (S2′, indicated by the arrow. B) Effect of pan-lysosomal protease inhibitor (CPI) on MHV and MHV-S2′FCS infection. HeLa-mCC1a cells were pretreated with CPI for 30 min and inoculated at MOI = 0.2 with luciferase expression cassette containing MHV-EFLM or MHV-S2′FCS in the presence of CPI, after which incubations were continued in the presence of CPI until 7 hpi. Infection levels were determined by measuring the luciferase activity in cell lysates relative to mock-treated cells. Error bars represent SEM, n = 3*3.
Figure 8. Furin inhibitor reduces infection with…
Figure 8. Furin inhibitor reduces infection with MHV-S2′FCS and renders the virus sensitive to endosomal maturation.
Haploid HAP1 cells (HAP1), haploid cells lacking VPS33A (H1-ΔV33) or VPS33A-lacking haploid cells retransfected with FLAG-tagged VLP33A (H1-ΔV33-fV33) were infected (MOI = 0.2) with MHV-EFLM (MHV-wt) or MHV-S2′FCS for 7 h. Where indicated, cells were treated with furin inhibitor (FI). Infection levels were determined by measuring the luciferase activity in cell lysates relative to mock-treated cells. Error bars represent SEM, n = 3*3.
Figure 9. MHV-S2′FCS fuses in early endosomes.
Figure 9. MHV-S2′FCS fuses in early endosomes.
siRNA-mediated gene silencing was performed as described in the legend to Figure 1. At 72 h post transfection, HeLa-mCC1a were inoculated with MHV-EFLM or MHV-S2′FCS at MOI = 0.2 and incubated until 7 hpi. Infection levels were determined by measuring the luciferase activity in cell lysates relative to mock-treated cells. Dotted line shows the lower 95% confidence interval of the negative siRNA controls. Error bars represent SEM, n = 3*3.
Figure 10. Entry of FIPV.
Figure 10. Entry of FIPV.
A) Clustal W alignment of spike proteins from several coronaviruses. Displayed is the fusion peptide (boxed) and the area upstream thereof. The area immediately upstream of the fusion peptide that contains the optimal FCS site (RRRRR) in MHV-S2′FCS is also boxed. B) siRNA-mediated gene silencing was performed as described in the legend to Figure 1. At 72 h post transfection, HeLa-fAPN cells were inoculated at MOI = 0.2 with luciferase expressing FIPV-RLuc. At 7 hpi infection was determined by measuring the luciferase activity in cell lysates and displayed relative to mock treated infection (inf). Error bars represent SEM, n = 3*3. Dotted line shows the lower 95% confidence interval of the negative siRNA controls. C) HeLa-fAPN cells inoculated with FIPV-Rluc at MOI = 0.1 were treated with pan-lysosomal protease inhibitor (CPI) or furin inhibitor (FI) from 30 min prior to 7 h post inoculation (0–7 h) or from 2–7 h post inoculation (2–7 h; hatched bars). Infection levels were determined by measuring the luciferase activity in cell lysates relative to mock-treated cells. Error bars represent SEM, n = 3*3.
Figure 11. MERS-CoV requires cleavage by furin…
Figure 11. MERS-CoV requires cleavage by furin but not by lysosomal proteases for infection.
Huh-7 cells inoculated with MERS-CoV were treated with furin inhibitor (FI) or pan-lysosomal protease inhibitor (CPI) starting from 30 min prior to inoculation. Numbers of infected cells was determined by immunocytochemical staining. Error bars represent SEM, n = 3.
Figure 12. Model of early and late…
Figure 12. Model of early and late coronavirus fusion.
MHV and MHV-S2′FCS are taken up by DAB2-dependent clathrin-mediated endocytosis to end up in RAB5-containing early endosomes. The FCS of MHV-S2′FCS is cleaved by furin or furin-like enzymes to allow fusion of the virus in early endosomes. Trafficking of MHV from late endosomes to lysosomes (RAB7/LAMP1-positive compartments) is required for processing of MHV by lysosomal proteases and viral fusion to occur. We propose that the sequence immediately upstream of the FP is a key determinant of the intracellular site of fusion. MERS-CoV and FIPV enter cells via fusion in early endosomes or lysosomes, respectively. MERS-CoV, which contains a minimal FCS, is inhibited by furin inhibitor (FI) but not by the pan-lysosomal protease inhibitor (CPI). The opposite holds true for FIPV. Based on this model, we predict that IBV strain Beaudette and HCoV-NL63, which contain FCSs (Fig. 10), to fuse in early endosomes in a furin-dependent manner. Other CoVs that do not contain a FCS at this position are predicted to fuse in lysosomes.

References

    1. Fuller AO, Spear PG (1987) Anti-glycoprotein D antibodies that permit adsorption but block infection by herpes simplex virus 1 prevent virion-cell fusion at the cell surface. Proceedings of the National Academy of Sciences of the United States of America 84: 5454–5458.
    1. Sodeik B, Ebersold MW, Helenius A (1997) Microtubule-mediated transport of incoming herpes simplex virus 1 capsids to the nucleus. The Journal of cell biology 136: 1007–1021.
    1. Okada Y (1969) Factors in fusion of cells by HVJ. Current topics in microbiology and immunology 48: 102–128.
    1. Permanyer M, Ballana E, Este JA (2010) Endocytosis of HIV: anything goes. Trends in microbiology 18: 543–551.
    1. Stein BS, Gowda SD, Lifson JD, Penhallow RC, Bensch KG, et al. (1987) pH-independent HIV entry into CD4-positive T cells via virus envelope fusion to the plasma membrane. Cell 49: 659–668.
    1. Authier F, Posner BI, Bergeron JJ (1996) Endosomal proteolysis of internalized proteins. FEBS letters 389: 55–60.
    1. Huotari J, Helenius A (2011) Endosome maturation. The EMBO journal 30: 3481–3500.
    1. Plemper RK (2011) Cell entry of enveloped viruses. Current opinion in virology 1: 92–100.
    1. Sieczkarski SB, Whittaker GR (2003) Differential requirements of Rab5 and Rab7 for endocytosis of influenza and other enveloped viruses. Traffic 4: 333–343.
    1. Skehel JJ, Bayley PM, Brown EB, Martin SR, Waterfield MD, et al. (1982) Changes in the conformation of influenza virus hemagglutinin at the pH optimum of virus-mediated membrane fusion. Proceedings of the National Academy of Sciences of the United States of America 79: 968–972.
    1. Carneiro FA, Ferradosa AS, Da Poian AT (2001) Low pH-induced conformational changes in vesicular stomatitis virus glycoprotein involve dramatic structure reorganization. The Journal of biological chemistry 276: 62–67.
    1. White J, Matlin K, Helenius A (1981) Cell fusion by Semliki Forest, influenza, and vesicular stomatitis viruses. The Journal of cell biology 89: 674–679.
    1. Krzyzaniak MA, Zumstein MT, Gerez JA, Picotti P, Helenius A (2013) Host cell entry of respiratory syncytial virus involves macropinocytosis followed by proteolytic activation of the F protein. PLoS pathogens 9: e1003309.
    1. Wool-Lewis RJ, Bates P (1999) Endoproteolytic processing of the ebola virus envelope glycoprotein: cleavage is not required for function. Journal of virology 73: 1419–1426.
    1. Zimmer G, Budz L, Herrler G (2001) Proteolytic activation of respiratory syncytial virus fusion protein. Cleavage at two furin consensus sequences. The Journal of biological chemistry 276: 31642–31650.
    1. Chandran K, Sullivan NJ, Felbor U, Whelan SP, Cunningham JM (2005) Endosomal proteolysis of the Ebola virus glycoprotein is necessary for infection. Science 308: 1643–1645.
    1. Peiris JS, Lai ST, Poon LL, Guan Y, Yam LY, et al. (2003) Coronavirus as a possible cause of severe acute respiratory syndrome. Lancet 361: 1319–1325.
    1. Zaki AM, van Boheemen S, Bestebroer TM, Osterhaus AD, Fouchier RA (2012) Isolation of a novel coronavirus from a man with pneumonia in Saudi Arabia. N Engl J Med 367: 1814–1820.
    1. de Haan CA, Rottier PJ (2005) Molecular interactions in the assembly of coronaviruses. Advances in virus research 64: 165–230.
    1. Bosch BJ, van der Zee R, de Haan CA, Rottier PJ (2003) The coronavirus spike protein is a class I virus fusion protein: structural and functional characterization of the fusion core complex. Journal of virology 77: 8801–8811.
    1. Inoue Y, Tanaka N, Tanaka Y, Inoue S, Morita K, et al. (2007) Clathrin-dependent entry of severe acute respiratory syndrome coronavirus into target cells expressing ACE2 with the cytoplasmic tail deleted. Journal of virology 81: 8722–8729.
    1. Wang H, Yang P, Liu K, Guo F, Zhang Y, et al. (2008) SARS coronavirus entry into host cells through a novel clathrin- and caveolae-independent endocytic pathway. Cell research 18: 290–301.
    1. Regan AD, Shraybman R, Cohen RD, Whittaker GR (2008) Differential role for low pH and cathepsin-mediated cleavage of the viral spike protein during entry of serotype II feline coronaviruses. Veterinary microbiology 132: 235–248.
    1. Van Hamme E, Dewerchin HL, Cornelissen E, Verhasselt B, Nauwynck HJ (2008) Clathrin- and caveolae-independent entry of feline infectious peritonitis virus in monocytes depends on dynamin. The Journal of general virology 89: 2147–2156.
    1. Nomura R, Kiyota A, Suzaki E, Kataoka K, Ohe Y, et al. (2004) Human coronavirus 229E binds to CD13 in rafts and enters the cell through caveolae. Journal of virology 78: 8701–8708.
    1. Eifart P, Ludwig K, Bottcher C, de Haan CA, Rottier PJ, et al. (2007) Role of endocytosis and low pH in murine hepatitis virus strain A59 cell entry. Journal of virology 81: 10758–10768.
    1. Qiu Z, Hingley ST, Simmons G, Yu C, Das Sarma J, et al. (2006) Endosomal proteolysis by cathepsins is necessary for murine coronavirus mouse hepatitis virus type 2 spike-mediated entry. Journal of virology 80: 5768–5776.
    1. Stauber R, Pfleiderera M, Siddell S (1993) Proteolytic cleavage of the murine coronavirus surface glycoprotein is not required for fusion activity. The Journal of general virology 74 (Pt 2) 183–191.
    1. Sturman LS, Ricard CS, Holmes KV (1985) Proteolytic cleavage of the E2 glycoprotein of murine coronavirus: activation of cell-fusing activity of virions by trypsin and separation of two different 90K cleavage fragments. Journal of virology 56: 904–911.
    1. de Haan CA, Stadler K, Godeke GJ, Bosch BJ, Rottier PJ (2004) Cleavage inhibition of the murine coronavirus spike protein by a furin-like enzyme affects cell-cell but not virus-cell fusion. Journal of virology 78: 6048–6054.
    1. Frana MF, Behnke JN, Sturman LS, Holmes KV (1985) Proteolytic cleavage of the E2 glycoprotein of murine coronavirus: host-dependent differences in proteolytic cleavage and cell fusion. Journal of virology 56: 912–920.
    1. Luytjes W, Sturman LS, Bredenbeek PJ, Charite J, van der Zeijst BA, et al. (1987) Primary structure of the glycoprotein E2 of coronavirus MHV-A59 and identification of the trypsin cleavage site. Virology 161: 479–487.
    1. Ricard CS, Sturman LS (1985) Isolation of the subunits of the coronavirus envelope glycoprotein E2 by hydroxyapatite high-performance liquid chromatography. Journal of chromatography 326: 191–197.
    1. Gombold JL, Hingley ST, Weiss SR (1993) Fusion-defective mutants of mouse hepatitis virus A59 contain a mutation in the spike protein cleavage signal. Journal of virology 67: 4504–4512.
    1. Leparc-Goffart I, Hingley ST, Chua MM, Jiang X, Lavi E, et al. (1997) Altered pathogenesis of a mutant of the murine coronavirus MHV-A59 is associated with a Q159L amino acid substitution in the spike protein. Virology 239: 1–10.
    1. Matsuyama S, Taguchi F (2009) Two-step conformational changes in a coronavirus envelope glycoprotein mediated by receptor binding and proteolysis. Journal of virology 83: 11133–11141.
    1. Simmons G, Gosalia DN, Rennekamp AJ, Reeves JD, Diamond SL, et al. (2005) Inhibitors of cathepsin L prevent severe acute respiratory syndrome coronavirus entry. Proceedings of the National Academy of Sciences of the United States of America 102: 11876–11881.
    1. Bosch BJ, Bartelink W, Rottier PJ (2008) Cathepsin L functionally cleaves the severe acute respiratory syndrome coronavirus class I fusion protein upstream of rather than adjacent to the fusion peptide. Journal of virology 82: 8887–8890.
    1. Simmons G, Reeves JD, Rennekamp AJ, Amberg SM, Piefer AJ, et al. (2004) Characterization of severe acute respiratory syndrome-associated coronavirus (SARS-CoV) spike glycoprotein-mediated viral entry. Proceedings of the National Academy of Sciences of the United States of America 101: 4240–4245.
    1. Belouzard S, Chu VC, Whittaker GR (2009) Activation of the SARS coronavirus spike protein via sequential proteolytic cleavage at two distinct sites. Proceedings of the National Academy of Sciences of the United States of America 106: 5871–5876.
    1. Matsuyama S, Ujike M, Morikawa S, Tashiro M, Taguchi F (2005) Protease-mediated enhancement of severe acute respiratory syndrome coronavirus infection. Proceedings of the National Academy of Sciences of the United States of America 102: 12543–12547.
    1. Belouzard S, Madu I, Whittaker GR (2010) Elastase-mediated activation of the severe acute respiratory syndrome coronavirus spike protein at discrete sites within the S2 domain. The Journal of biological chemistry 285: 22758–22763.
    1. Kam YW, Okumura Y, Kido H, Ng LF, Bruzzone R, et al. (2009) Cleavage of the SARS coronavirus spike glycoprotein by airway proteases enhances virus entry into human bronchial epithelial cells in vitro. PloS one 4: e7870.
    1. Bertram S, Glowacka I, Muller MA, Lavender H, Gnirss K, et al. (2011) Cleavage and activation of the severe acute respiratory syndrome coronavirus spike protein by human airway trypsin-like protease. Journal of virology 85: 13363–13372.
    1. Shulla A, Heald-Sargent T, Subramanya G, Zhao J, Perlman S, et al. (2011) A transmembrane serine protease is linked to the severe acute respiratory syndrome coronavirus receptor and activates virus entry. Journal of virology 85: 873–882.
    1. Yamada Y, Liu DX (2009) Proteolytic activation of the spike protein at a novel RRRR/S motif is implicated in furin-dependent entry, syncytium formation, and infectivity of coronavirus infectious bronchitis virus in cultured cells. Journal of virology 83: 8744–8758.
    1. Matthews SP, Werber I, Deussing J, Peters C, Reinheckel T, et al. (2010) Distinct protease requirements for antigen presentation in vitro and in vivo. Journal of immunology 184: 2423–2431.
    1. Watanabe R, Matsuyama S, Shirato K, Maejima M, Fukushi S, et al. (2008) Entry from the cell surface of severe acute respiratory syndrome coronavirus with cleaved S protein as revealed by pseudotype virus bearing cleaved S protein. Journal of virology 82: 11985–11991.
    1. Wicht O, Burkard C, de Haan CA, van Kuppeveld FJ, Rottier PJ, et al. (2014) Identification and characterization of a proteolytically primed form of the murine coronavirus spike proteins after fusion with the target cell. Journal of virology 88: 4943–4952.
    1. Snijder B, Sacher R, Ramo P, Liberali P, Mench K, et al. (2012) Single-cell analysis of population context advances RNAi screening at multiple levels. Molecular systems biology 8: 579.
    1. Gouin E, Welch MD, Cossart P (2005) Actin-based motility of intracellular pathogens. Current opinion in microbiology 8: 35–45.
    1. May RC (2001) The Arp2/3 complex: a central regulator of the actin cytoskeleton. Cellular and molecular life sciences: CMLS 58: 1607–1626.
    1. Pfeffer SR (2013) Rab GTPase regulation of membrane identity. Current opinion in cell biology 25: 414–419.
    1. Balderhaar HJ, Ungermann C (2013) CORVET and HOPS tethering complexes - coordinators of endosome and lysosome fusion. Journal of cell science 126: 1307–1316.
    1. Bonifacino JS, Hurley JH (2008) Retromer. Current opinion in cell biology 20: 427–436.
    1. DeMali KA, Burridge K (2003) Coupling membrane protrusion and cell adhesion. Journal of cell science 116: 2389–2397.
    1. Bagrodia S, Cerione RA (1999) Pak to the future. Trends in cell biology 9: 350–355.
    1. Robinson LJ, Aniento F, Gruenberg J (1997) NSF is required for transport from early to late endosomes. Journal of cell science 110 (Pt 17) 2079–2087.
    1. de Haan CA, van Genne L, Stoop JN, Volders H, Rottier PJ (2003) Coronaviruses as vectors: position dependence of foreign gene expression. Journal of virology 77: 11312–11323.
    1. Verheije MH, Raaben M, Mari M, Te Lintelo EG, Reggiori F, et al. (2008) Mouse hepatitis coronavirus RNA replication depends on GBF1-mediated ARF1 activation. PLoS pathogens 4: e1000088.
    1. Raaben M, Posthuma CC, Verheije MH, te Lintelo EG, Kikkert M, et al. (2010) The ubiquitin-proteasome system plays an important role during various stages of the coronavirus infection cycle. Journal of virology 84: 7869–7879.
    1. Huynh KK, Gershenzon E, Grinstein S (2008) Cholesterol accumulation by macrophages impairs phagosome maturation. The Journal of biological chemistry 283: 35745–35755.
    1. Burkard C, Bloyet LM, Wicht O, van Kuppeveld FJ, Rottier PJ, et al. (2014) Dissecting Virus Entry: Replication-Independent Analysis of Virus Binding, Internalization, and Penetration Using Minimal Complementation of beta-Galactosidase. PloS one 9: e101762.
    1. Langley KE, Villarejo MR, Fowler AV, Zamenhof PJ, Zabin I (1975) Molecular basis of beta-galactosidase alpha-complementation. Proceedings of the National Academy of Sciences of the United States of America 72: 1254–1257.
    1. Engel S, Heger T, Mancini R, Herzog F, Kartenbeck J, et al. (2011) Role of endosomes in simian virus 40 entry and infection. Journal of virology 85: 4198–4211.
    1. Bayer N, Schober D, Prchla E, Murphy RF, Blaas D, et al. (1998) Effect of bafilomycin A1 and nocodazole on endocytic transport in HeLa cells: implications for viral uncoating and infection. Journal of virology 72: 9645–9655.
    1. Johannsdottir HK, Mancini R, Kartenbeck J, Amato L, Helenius A (2009) Host cell factors and functions involved in vesicular stomatitis virus entry. Journal of virology 83: 440–453.
    1. Le Blanc I, Luyet PP, Pons V, Ferguson C, Emans N, et al. (2005) Endosome-to-cytosol transport of viral nucleocapsids. Nature cell biology 7: 653–664.
    1. Matos PM, Marin M, Ahn B, Lam W, Santos NC, et al. (2013) Anionic lipids are required for vesicular stomatitis virus G protein-mediated single particle fusion with supported lipid bilayers. The Journal of biological chemistry 288: 12416–12425.
    1. Skehel JJ, Wiley DC (2000) Receptor binding and membrane fusion in virus entry: the influenza hemagglutinin. Annual review of biochemistry 69: 531–569.
    1. Tani H, Komoda Y, Matsuo E, Suzuki K, Hamamoto I, et al. (2007) Replication-competent recombinant vesicular stomatitis virus encoding hepatitis C virus envelope proteins. Journal of virology 81: 8601–8612.
    1. Whitt MA (2010) Generation of VSV pseudotypes using recombinant DeltaG-VSV for studies on virus entry, identification of entry inhibitors, and immune responses to vaccines. Journal of virological methods 169: 365–374.
    1. Konig R, Stertz S, Zhou Y, Inoue A, Hoffmann HH, et al. (2010) Human host factors required for influenza virus replication. Nature 463: 813–817.
    1. Carette JE, Raaben M, Wong AC, Herbert AS, Obernosterer G, et al. (2011) Ebola virus entry requires the cholesterol transporter Niemann-Pick C1. Nature 477: 340–343.
    1. Tveten K, Ranheim T, Berge KE, Leren TP, Kulseth MA (2009) The effect of bafilomycin A1 and protease inhibitors on the degradation and recycling of a Class 5-mutant LDLR. Acta biochimica et biophysica Sinica 41: 246–255.
    1. van Kasteren SI, Berlin I, Colbert JD, Keane D, Ovaa H, et al. (2011) A multifunctional protease inhibitor to regulate endolysosomal function. ACS chemical biology 6: 1198–1204.
    1. White JM, Delos SE, Brecher M, Schornberg K (2008) Structures and mechanisms of viral membrane fusion proteins: multiple variations on a common theme. Critical reviews in biochemistry and molecular biology 43: 189–219.
    1. Thomas G (2002) Furin at the cutting edge: from protein traffic to embryogenesis and disease. Nature reviews Molecular cell biology 3: 753–766.
    1. de Haan CA, Haijema BJ, Boss D, Heuts FW, Rottier PJ (2005) Coronaviruses as vectors: stability of foreign gene expression. Journal of virology 79: 12742–12751.
    1. Gallagher TM (1997) A role for naturally occurring variation of the murine coronavirus spike protein in stabilizing association with the cellular receptor. Journal of virology 71: 3129–3137.
    1. Krueger DK, Kelly SM, Lewicki DN, Ruffolo R, Gallagher TM (2001) Variations in disparate regions of the murine coronavirus spike protein impact the initiation of membrane fusion. Journal of virology 75: 2792–2802.
    1. Phillips JJ, Chua MM, Lavi E, Weiss SR (1999) Pathogenesis of chimeric MHV4/MHV-A59 recombinant viruses: the murine coronavirus spike protein is a major determinant of neurovirulence. Journal of virology 73: 7752–7760.
    1. Pu Y, Zhang X (2008) Mouse hepatitis virus type 2 enters cells through a clathrin-mediated endocytic pathway independent of Eps15. Journal of virology 82: 8112–8123.
    1. Choi KS, Aizaki H, Lai MM (2005) Murine coronavirus requires lipid rafts for virus entry and cell-cell fusion but not for virus release. Journal of virology 79: 9862–9871.
    1. Thorp EB, Gallagher TM (2004) Requirements for CEACAMs and cholesterol during murine coronavirus cell entry. Journal of virology 78: 2682–2692.
    1. Fretz M, Jin J, Conibere R, Penning NA, Al-Taei S, et al. (2006) Effects of Na+/H+ exchanger inhibitors on subcellular localisation of endocytic organelles and intracellular dynamics of protein transduction domains HIV-TAT peptide and octaarginine. Journal of controlled release: official journal of the Controlled Release Society 116: 247–254.
    1. Lagana A, Vadnais J, Le PU, Nguyen TN, Laprade R, et al. (2000) Regulation of the formation of tumor cell pseudopodia by the Na(+)/H(+) exchanger NHE1. Journal of cell science 113 (Pt 20) 3649–3662.
    1. Meier O, Boucke K, Hammer SV, Keller S, Stidwill RP, et al. (2002) Adenovirus triggers macropinocytosis and endosomal leakage together with its clathrin-mediated uptake. The Journal of cell biology 158: 1119–1131.
    1. Wadia JS, Stan RV, Dowdy SF (2004) Transducible TAT-HA fusogenic peptide enhances escape of TAT-fusion proteins after lipid raft macropinocytosis. Nature medicine 10: 310–315.
    1. Ivanov AI, Nusrat A, Parkos CA (2004) Endocytosis of epithelial apical junctional proteins by a clathrin-mediated pathway into a unique storage compartment. Molecular biology of the cell 15: 176–188.
    1. Kaksonen M, Toret CP, Drubin DG (2006) Harnessing actin dynamics for clathrin-mediated endocytosis. Nature reviews Molecular cell biology 7: 404–414.
    1. Huang IC, Bosch BJ, Li F, Li W, Lee KH, et al. (2006) SARS coronavirus, but not human coronavirus NL63, utilizes cathepsin L to infect ACE2-expressing cells. The Journal of biological chemistry 281: 3198–3203.
    1. Lee DH, Goldberg AL (1998) Proteasome inhibitors: valuable new tools for cell biologists. Trends in cell biology 8: 397–403.
    1. Tawa NE Jr, Odessey R, Goldberg AL (1997) Inhibitors of the proteasome reduce the accelerated proteolysis in atrophying rat skeletal muscles. The Journal of clinical investigation 100: 197–203.
    1. van Kerkhof P, Alves dos Santos CM, Sachse M, Klumperman J, Bu G, et al. (2001) Proteasome inhibitors block a late step in lysosomal transport of selected membrane but not soluble proteins. Molecular biology of the cell 12: 2556–2566.
    1. Zaarur N, Meriin AB, Bejarano E, Xu X, Gabai VL, et al. (2014) Proteasome failure promotes positioning of lysosomes around the aggresome via local block of microtubule-dependent transport. Molecular and cellular biology 34: 1336–1348.
    1. Belouzard S, Millet JK, Licitra BN, Whittaker GR (2012) Mechanisms of coronavirus cell entry mediated by the viral spike protein. Viruses 4: 1011–1033.
    1. Kuo L, Godeke GJ, Raamsman MJ, Masters PS, Rottier PJ (2000) Retargeting of coronavirus by substitution of the spike glycoprotein ectodomain: crossing the host cell species barrier. J Virol 74: 1393–1406.
    1. van Boheemen S, de Graaf M, Lauber C, Bestebroer TM, Raj VS, et al. (2012) Genomic characterization of a newly discovered coronavirus associated with acute respiratory distress syndrome in humans. mBio 3.
    1. Bosch BJ, van der Zee R, de Haan CA, Rottier PJ (2003) The coronavirus spike protein is a class I virus fusion protein: structural and functional characterization of the fusion core complex. J Virol 77: 8801–8811.
    1. Dveksler GS, Pensiero MN, Cardellichio CB, Williams RK, Jiang GS, et al. (1991) Cloning of the mouse hepatitis virus (MHV) receptor: expression in human and hamster cell lines confers susceptibility to MHV. Journal of virology 65: 6881–6891.
    1. de Haan CA, Haijema BJ, Masters PS, Rottier PJ (2008) Manipulation of the coronavirus genome using targeted RNA recombination with interspecies chimeric coronaviruses. Methods Mol Biol 454: 229–236.
    1. Kuo L, Godeke GJ, Raamsman MJ, Masters PS, Rottier PJ (2000) Retargeting of coronavirus by substitution of the spike glycoprotein ectodomain: crossing the host cell species barrier. Journal of virology 74: 1393–1406.
    1. Vonderheit A, Helenius A (2005) Rab7 associates with early endosomes to mediate sorting and transport of Semliki forest virus to late endosomes. PLoS biology 3: e233.
    1. Schulze H, Kolter T, Sandhoff K (2009) Principles of lysosomal membrane degradation: Cellular topology and biochemistry of lysosomal lipid degradation. Biochimica et biophysica acta 1793: 674–683.

Source: PubMed

3
订阅