Glycopeptide Antibiotics Potently Inhibit Cathepsin L in the Late Endosome/Lysosome and Block the Entry of Ebola Virus, Middle East Respiratory Syndrome Coronavirus (MERS-CoV), and Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV)

Nan Zhou, Ting Pan, Junsong Zhang, Qianwen Li, Xue Zhang, Chuan Bai, Feng Huang, Tao Peng, Jianhua Zhang, Chao Liu, Liang Tao, Hui Zhang, Nan Zhou, Ting Pan, Junsong Zhang, Qianwen Li, Xue Zhang, Chuan Bai, Feng Huang, Tao Peng, Jianhua Zhang, Chao Liu, Liang Tao, Hui Zhang

Abstract

Ebola virus infection can cause severe hemorrhagic fever with a high mortality in humans. The outbreaks of Ebola viruses in 2014 represented the most serious Ebola epidemics in history and greatly threatened public health worldwide. The development of additional effective anti-Ebola therapeutic agents is therefore quite urgent. In this study, via high throughput screening of Food and Drug Administration-approved drugs, we identified that teicoplanin, a glycopeptide antibiotic, potently prevents the entry of Ebola envelope pseudotyped viruses into the cytoplasm. Furthermore, teicoplanin also has an inhibitory effect on transcription- and replication-competent virus-like particles, with an IC50 as low as 330 nm Comparative analysis further demonstrated that teicoplanin is able to block the entry of Middle East respiratory syndrome (MERS) and severe acute respiratory syndrome (SARS) envelope pseudotyped viruses as well. Teicoplanin derivatives such as dalbavancin, oritavancin, and telavancin can also inhibit the entry of Ebola, MERS, and SARS viruses. Mechanistic studies showed that teicoplanin blocks Ebola virus entry by specifically inhibiting the activity of cathepsin L, opening a novel avenue for the development of additional glycopeptides as potential inhibitors of cathepsin L-dependent viruses. Notably, given that teicoplanin has routinely been used in the clinic with low toxicity, our work provides a promising prospect for the prophylaxis and treatment of Ebola, MERS, and SARS virus infection.

Keywords: Ebola virus; MERS-CoV; SARS-CoV; antibiotics; glycopeptide; glycoprotein; lysosome; virus entry.

© 2016 by The American Society for Biochemistry and Molecular Biology, Inc.

Figures

FIGURE 1.
FIGURE 1.
Teicoplanin specifically inhibits the entry of Ebola viruses.A, teicoplanin and amiodarone specifically inhibit the entry of Ebola viruses. HEK293T cells were seeded in a 96-well plate, and 24 h later, the cells were incubated with various reagents at 37 °C for 1 h. The cells were then infected with HIV-luc/Zaire EBOV-GP (2014) pseudotyped viruses. After washing and incubation with fresh medium for 48 h, the intracellular luciferase activity was measured.B, chemical structure of teicoplanin.C, HEK293T cells were incubated with teicoplanin at various concentrations at 37 °C for 1 h. The intracellular luciferase activity was measured at 48 h post-infection. The IC50 was calculated using GraphPad Prism software. D, HEK293T cells were incubated with 500 μm teicoplanin at 37 °C for 48 h. Then the cell viability was determined.E, HEK293T cells were infected with HIV-luc/Zaire EBOV-GP (2014) pseudotyped viruses and incubated with 50 μm teicoplanin at 0, 2, 4, and 8 h post-infection. The cells were then incubated for 48 h after which the intracellular luciferase activity was tested. The HIV-luc/VSV-G pseudotyped viruses were used as the controls for specificity. F, HEK293T cells were seeded in a 6-well plate (2 × 105 per well). After 24 h, the cells were incubated with teicoplanin at various concentrations at 37 °C for 1 h and then infected with p24-normalized (100 ng) HIV-luc/Zaire EBOV-GP (2014) or HIV-luc/VSV-G pseudotyped viruses per well. After 6 h, the cells were washed with PBS twice and incubated with 0.25% trypsin at 37 °C for 2 min to remove the viruses that adhered to the cell surfaces. The cells were then collected and lysed, and the intracellular amount of HIV-1 p24 was measured by ELISA. The results are representatives of at least three independent experiments. Thebars show the mean values ± S.D. (error bars). The p value was determined by a Student's t test. ***,p < 0.001.
FIGURE 2.
FIGURE 2.
Ebola virus entry into different cell types is repressed by teicoplanin.A, primary human umbilical vein endothelial cells were seeded in a 96-well plate. After 24 h, the cells were incubated with teicoplanin at various concentrations at 37 °C for 1 h. Subsequently, the cells were infected with HIV-luc/Zaire EBOV-GP (2014) pseudotyped viruses. After washing and incubation with fresh medium for 48 h, the intracellular luciferase activity was measured. B, antiviral entry assay of teicoplanin on the A549 cells was conducted in a similar way. C, antiviral entry assay of teicoplanin on the HeLa cells was conducted in a similar procedure. D, THP-1 cells were incubated with teicoplanin at various concentrations at 37 °C for 1 h. Subsequently, the cells were infected with HIV-luc/Zaire EBOV-GP (2014) pseudotyped viruses. After 12 h, the supernatants were removed by centrifugation at 300 × g for 10 min, and the cells were suspended and cultured with RPMI 1640 medium at 37 °C for 48 h. The intracellular luciferase activity was then tested. The results are representative of at least three independent experiments. The barsshow the mean values ± S.D. (error bars). The p value was determined by a Student'st test. **, p < 0.01; ***, p < 0.001.
FIGURE 3.
FIGURE 3.
Target of teicoplanin is located within the host cells.A, p24-normalized (50 ng) HIV-luc/Zaire EBOV-GP (2014) pseudotyped viruses were incubated with 50 μm teicoplanin in a 96-well plate at 37 °C for 12 h. Then the compound virus mixtures were transferred into a Microcon 30-kDa centrifugal filter device (Millipore) and centrifuged (7000 × g) at 4 °C for 15 min. Subsequently, fresh medium was added twice onto the filter device to wash the compound virus mixtures. Then 0.5 ml of DMEM was used to suspend the compound virus mixtures, and the reversed filter device was centrifuged (500 ×g) at 4 °C for 5 min. The solution was collected and used to infect HEK293T cells after HIV-1 p24 normalization. After 48 h, the intracellular luciferase activity was measured. B, HEK293T cells were incubated with teicoplanin at various concentrations for 12 h. The cells were then infected with HIV-luc/Zaire EBOV-GP (2014) pseudotyped viruses. After washing and incubation with fresh medium for 48 h, the intracellular luciferase activity was measured. The assay during treatment was the same as the antiviral entry assay. The results are representative of at least three independent experiments. The bars show the mean values ± S.D. (error bars). Thep value was determined by a Student'st test. n.s., not significant; **, p < 0.01; ***,p < 0.001.
FIGURE 4.
FIGURE 4.
Teicoplanin does not block the cell receptor.A, schematic representation of the entry of Ebola viruses and SARS-CoVs. B, HEK293T cells were incubated with teicoplanin at various concentrations at 37 °C for 1 h. The cells were then infected with HIV-luc/SARS-CoV-S pseudotyped viruses. After washing and incubation with fresh medium for 48 h, the intracellular luciferase activity was measured. C, HEK293T cells were seeded in a 96-well plate. After 24 h, the cells were transfected with 200 nm siRNAs against NPC1 and ACE2, respectively. After 48 h, the cells were infected with HIV-luc/Zaire EBOV-GP (2014) or HIV-luc/SARS-CoV-S pseudotyped viruses. The results are representative of at least three independent experiments. The bars show the mean values ± S.D. (error bars). Thep value was determined by a Student'st test. n.s., not significant; *, p < 0.05; **,p < 0.01; ***, p < 0.001.
FIGURE 5.
FIGURE 5.
Teicoplanin has no effect on HOPS complexes.A, schematic representation of the entry of Ebola viruses, SARS-CoVs, and VSVs. The host factors thatare essential for the entry of Ebola viruses are illustrated.B, HEK293T cells were transfected with 200 nm siRNAs per well. After 48 h, the cells were infected with HIV-luc/Zaire EBOV-GP (2014), HIV-luc/SARS-CoV-S, or HIV-luc/VSV pseudotyped viruses. After washing and incubation with fresh medium for 48 h, the intracellular luciferase activity was measured. C, HEK293T cells were plated in a glass bottom dish. After 24 h, groups of cells were incubated with 50 μm teicoplanin or control for 12 h. Another group of cells was transfected with siRNAs against VPS39 and VPS41 or the si-control and incubated with these siRNAs at a final concentration of 200 nm for 48 h. Then the cells were incubated with dextran-Alexa Flour 568 (Mr 10,000) for 2 h in 1% serum-containing DMEM. Subsequently, the cells were subjected to immunofluorescence analysis. Images were obtained using Zeiss LSM780 confocal microscopy with Zeiss ZFN software.Scale bar, 10 μm. D, co-localization of dextran and LAMP1 was analyzed from 20 fields (≥5 cells per field) per independent experiment. Pearson's overlap coefficients were used to determine the levels of the co-localization of dextran and LAMP1. Eand F, HEK293T cells were incubated with teicoplanin at the various indicated concentrations at 37 °C for 1 h. The cells were then infected with HIV-luc/Zaire EBOV-GP (2014) or HIV-luc/VSV-G pseudotyped viruses, respectively. Subsequently, the cells were incubated with 0.25% trypsin at 37 °C for 2 min to remove the viruses that adhered to the cell surfaces at 0.5, 1, and 2 h post-infection. The intracellular amount of HIV-1 p24 was then measured by ELISA. The results are representative of at least three independent experiments. The bars show the mean values ± S.D. (error bars). Thep value was determined by a Student'st test. n.s., not significant; ***, p < 0.001.
FIGURE 6.
FIGURE 6.
Teicoplanin directly inhibits the activity of cathepsin L.A, schematic representation of the cathepsin L enzymatic inhibition assay. B, HEK293T cells were incubated with teicoplanin or Z-Phe-Tyr(t-Bu)-diazomethyl ketones and then incubated with 50 μm (Z-Phe-Arg)2-R110 at 37 °C for 90 min. The level of fluorescence was determined using a fluorometer with excitation at 488 nm and emission at 510 nm. C, HEK293T cells were incubated with teicoplanin at various concentrations at 37 °C for 48 h. The cell viability was then determined using the CellTiter® 96 Aqueous One Solution Cell Proliferation Assay. D, HEK293T cells were incubated with teicoplanin or Z-Phe-Tyr(t-Bu)-diazomethyl ketones at 37 °C for 1 h. The cells were then infected with HIV-luc/Zaire EBOV-GP (2014) or HIV-luc/VSV-G pseudotyped viruses. After incubation for 48 h, the intracellular luciferase activity was measured. E, recombinant human cathepsin L (rhCTSL), teicoplanin, or Z-Phe-Tyr(t-Bu)-diazomethyl ketones were incubated 37 °C for 4 h. The rhCTSL-compound mixtures were incubated with (Z-Phe-Arg)2-R110 at 37 °C for 4 h. The fluorescence was then tested using a fluorometer with excitation at 488 nm and emission at 510 nm. The results are representative of at least three independent experiments. Thebars show the mean values ± S.D. (error bars). The p value was determined by a Student's t test.n.s., not significant, *,p < 0.05; **, p < 0.01; ***, p < 0.001.
FIGURE 7.
FIGURE 7.
Entry of Ebola trVLPs is repressed by glycopeptide antibiotics with the exception of vancomycin.A, schematic representation of the EbolatrVLPs entry assay. B, for the pre-transfection experiment, HEK293T cells were pre-transfected with 12.5 ng of pCAGGS-NP, 12.5 ng of pCAGGS-VP35, 7.5 ng of pCAGGS-VP30, 100 ng of pCAGGS-L, 25 ng of pCAGGS-T7, and 25 ng of pCAGGS-Tim1 plasmids. After 24 h, the medium was discarded, and the cells were incubated with teicoplanin at various concentrations at 37 °C for 1 h. After washing and incubation with fresh medium for 48 h, the intracellular luciferase activity was measured. The IC50 was calculated using GraphPad Prism software. C, for the non-pre-transfection experiment, HEK293T cells were incubated with teicoplanin at various concentrations at 37 °C for 1 h without pre-transfection of the above plasmids. The methods used for conducting Ebola trVLPs infection and measuring the intracellular luciferase activity were the same as those described in B. D and E, IC50 values of dalbavancin on the entry of Ebola trVLPs were determined under pre-transfection and non-pre-transfection conditions, respectively.F and G, IC50 values of oritavancin on the entry of Ebola trVLPs were calculated using pre-transfection or non-pre-transfection conditions, respectively. H andI, IC50 values of telavancin on the entry of Ebola trVLPs were determined under pre-transfection and non-pre-transfection conditions, respectively.J and K, effect of vancomycin on the entry of Ebola trVLPs was determined using pre-transfection or non-pre-transfection conditions, respectively. The results are representative of at least three independent experiments. The bars show the mean values ± S.D. (error bars). Thep value was determined by a Student'st test. n.s., not significant.
FIGURE 8.
FIGURE 8.
Entry of MERS-CoVs and SARS-CoVs is inhibited by glycopeptide antibiotics with the exception of vancomycin.A and E, HEK293T cells were incubated with teicoplanin at various concentrations at 37 °C for 1 h. The cells were infected with HIV-luc/MERS-CoV-S or HIV-luc/SARS-CoV-S pseudotyped viruses. After washing and incubation with fresh medium for 48 h, the intracellular luciferase activity was measured. The IC50 was calculated using GraphPad Prism software. B andF, IC50 values of dalbavancin on the entry of HIV-luc/MERS-CoV-S and HIV-luc/SARS-CoV-S pseudotyped viruses were determined. C andG, IC50 values of oritavancin on the entry of HIV-luc/MERS-CoV-S and HIV-luc/SARS-CoV-S pseudotyped viruses were calculated. D andH, IC50 values of telavancin on the entry of HIV-luc/MERS-CoV-S and HIV-luc/SARS-CoV-S pseudotyped viruses were determined. I andJ, The effect of vancomycin on the entry of HIV-luc/MERS-CoV-S and HIV-luc/SARS-CoV-S pseudotyped viruses was determined. The results are representative of at least three independent experiments. The bars show the mean values ± S.D. (error bars). Thep value was determined by a Student'st test. n.s., not significant.
FIGURE 9.
FIGURE 9.
Chemical structures of glycopeptide antibiotics. Teicoplanin, dalbavancin, oritavancin, and telavancin, which inhibit the entry of Ebola trVLPs, MERS-CoVs, and SARS-CoVs, contain key hydrophobic groups. However, vancomycin, which does not exert antiviral activity, does not contain these groups.

References

    1. Kiley M. P., Bowen E. T., Eddy G. A., Isaäcson M., Johnson K. M., McCormick J. B., Murphy F. A., Pattyn S. R., Peters D., Prozesky O. W., Regnery R. L., Simpson D. I., Slenczka W., Sureau P., van der Groen G., et al. (1982) Filoviridae: a taxonomic home for Marburg and Ebola viruses? Intervirology 18, 24–32
    1. World Health Organization (1978) Ebola haemorrhagic fever in Zaire, 1976. Bull. World Health Organ. 56, 271–293
    1. World Health Organization (1978) Ebola haemorrhagic fever in Sudan, 1976. Report of a WHO/International Study Team. Bull. World Health Organ. 56, 247–270
    1. Jahrling P. B., Geisbert T. W., Dalgard D. W., Johnson E. D., Ksiazek T. G., Hall W. C., and Peters C. J. (1990) Preliminary report: isolation of Ebola virus from monkeys imported to U.S.A. Lancet 335, 502–505
    1. Le Guenno B., Formenty P., Formentry P, Wyers M., Gounon P., Walker F., and Boesch C. (1995) Isolation and partial characterisation of a new strain of Ebola virus. Lancet 345, 1271–1274
    1. Towner J. S., Sealy T. K., Khristova M. L., Albariño C. G., Conlan S., Reeder S. A., Quan P. L., Lipkin W. I., Downing R., Tappero J. W., Okware S., Lutwama J., Bakamutumaho B., Kayiwa J., Comer J. A., et al. (2008) Newly discovered Ebola virus associated with hemorrhagic fever outbreak in Uganda. PLoS Pathog. 4, e1000212.
    1. Alvarez C. P., Lasala F., Carrillo J., Muñiz O., Corbí A. L., and Delgado R. (2002) C-type lectins DC-SIGN and L-SIGN mediate cellular entry by Ebola virus in cis and in trans. J. Virol. 76, 6841–6844
    1. Chan S. Y., Empig C. J., Welte F. J., Speck R. F., Schmaljohn A., Kreisberg J. F., and Goldsmith M. A. (2001) Folate receptor-α is a cofactor for cellular entry by Marburg and Ebola viruses. Cell 106, 117–126
    1. Gramberg T., Hofmann H., Möller P., Lalor P. F., Marzi A., Geier M., Krumbiegel M., Winkler T., Kirchhoff F., Adams D. H., Becker S., Münch J., and Pöhlmann S. (2005) LSECtin interacts with filovirus glycoproteins and the spike protein of SARS coronavirus. Virology 340, 224–236
    1. Kondratowicz A. S., Lennemann N. J., Sinn P. L., Davey R. A., Hunt C. L., Moller-Tank S., Meyerholz D. K., Rennert P., Mullins R. F., Brindley M., Sandersfeld L. M., Quinn K., Weller M., McCray P. B. Jr., Chiorini J., and Maury W. (2011) T-cell immunoglobulin and mucin domain 1 (TIM-1) is a receptor for Zaire Ebolavirus and Lake Victoria Marburgvirus. Proc. Natl. Acad. Sci. U.S.A. 108, 8426–8431
    1. O'Hearn A., Wang M., Cheng H., Lear-Rooney C. M., Koning K., Rumschlag-Booms E., Varhegyi E., Olinger G., and Rong L. (2015) Role of EXT1 and glycosaminoglycans in the early stage of filovirus entry. J. Virol. 89, 5441–5449
    1. Shimojima M., Takada A., Ebihara H., Neumann G., Fujioka K., Irimura T., Jones S., Feldmann H., and Kawaoka Y. (2006) Tyro3 family-mediated cell entry of Ebola and Marburg viruses. J. Virol. 80, 10109–10116
    1. Takada A., Watanabe S., Ito H., Okazaki K., Kida H., and Kawaoka Y. (2000) Downregulation of β1 integrins by Ebola virus glycoprotein: implication for virus entry. Virology 278, 20–26
    1. Saeed M. F., Kolokoltsov A. A., Albrecht T., and Davey R. A. (2010) Cellular entry of ebola virus involves uptake by a macropinocytosis-like mechanism and subsequent trafficking through early and late endosomes. PLoS Pathog. 6, e1001110.
    1. Nanbo A., Imai M., Watanabe S., Noda T., Takahashi K., Neumann G., Halfmann P., and Kawaoka Y. (2010) Ebolavirus is internalized into host cells via macropinocytosis in a viral glycoprotein-dependent manner. PLoS Pathog. 6, e1001121.
    1. Carette J. E., Raaben M., Wong A. C., Herbert A. S., Obernosterer G., Mulherkar N., Kuehne A. I., Kranzusch P. J., Griffin A. M., Ruthel G., Dal Cin P., Dye J. M., Whelan S. P., Chandran K., and Brummelkamp T. R. (2011) Ebola virus entry requires the cholesterol transporter Niemann-Pick C1. Nature 477, 340–343
    1. Chandran K., Sullivan N. J., Felbor U., Whelan S. P., and Cunningham J. M. (2005) Endosomal proteolysis of the Ebola virus glycoprotein is necessary for infection. Science 308, 1643–1645
    1. Mingo R. M., Simmons J. A., Shoemaker C. J., Nelson E. A., Schornberg K. L., D'Souza R. S., Casanova J. E., and White J. M. (2015) Ebola virus and severe acute respiratory syndrome coronavirus display late cell entry kinetics: evidence that transport to NPC1+ endolysosomes is a rate-defining step. J. Virol. 89, 2931–2943
    1. Rampling T., Ewer K., Bowyer G., Wright D., Imoukhuede E. B., Payne R., Hartnell F., Gibani M., Bliss C., Minhinnick A., Wilkie M., Venkatraman N., Poulton I., Lella N., Roberts R., et al. (2015) A monovalent chimpanzee adenovirus Ebola vaccine- preliminary report. N. Engl. J. Med. 10.1056/NEJMoa1411627
    1. Regules J. A., Beigel J. H., Paolino K. M., Voell J., Castellano A. R., Munoz P., Moon J. E., Ruck R. C., Bennett J. W., Twomey P. S., Gutierrez R. L., Remich S. A., Hack H. R., Wisniewski M. L., Josleyn M. D., et al. (2015) A recombinant vesicular stomatitis virus Ebola vaccine–preliminary report. N. Engl. J. Med. 10.1056/NEJMoa1414216
    1. Qiu X., Wong G., Audet J., Bello A., Fernando L., Alimonti J. B., Fausther-Bovendo H., Wei H., Aviles J., Hiatt E., Johnson A., Morton J., Swope K., Bohorov O., Bohorova N., et al. (2014) Reversion of advanced Ebola virus disease in nonhuman primates with ZMapp. Nature 514, 47–53
    1. Geisbert T. W., Lee A. C., Robbins M., Geisbert J. B., Honko A. N., Sood V., Johnson J. C., de Jong S., Tavakoli I., Judge A., Hensley L. E., and Maclachlan I. (2010) Postexposure protection of non-human primates against a lethal Ebola virus challenge with RNA interference: a proof-of-concept study. Lancet 375, 1896–1905
    1. Warren T. K., Wells J., Panchal R. G., Stuthman K. S., Garza N. L., Van Tongeren S. A., Dong L., Retterer C. J., Eaton B. P., Pegoraro G., Honnold S., Bantia S., Kotian P., Chen X., Taubenheim B. R., et al. (2014) Protection against filovirus diseases by a novel broad-spectrum nucleoside analog BCX4430. Nature 508, 402–405
    1. Wolf M. C., Freiberg A. N., Zhang T., Akyol-Ataman Z., Grock A., Hong P. W., Li J., Watson N. F., Fang A. Q., Aguilar H. C., Porotto M., Honko A. N., Damoiseaux R., Miller J. P., Woodson S. E., et al. (2010) A broad-spectrum antiviral targeting entry of enveloped viruses. Proc. Natl. Acad. Sci. U.S.A. 107, 3157–3162
    1. Gehring G., Rohrmann K., Atenchong N., Mittler E., Becker S., Dahlmann F., Pöhlmann S., Vondran F. W., David S., Manns M. P., Ciesek S., and von Hahn T. (2014) The clinically approved drugs amiodarone, dronedarone and verapamil inhibit filovirus cell entry. J. Antimicrob. Chemother. 69, 2123–2131
    1. Wang Y., Cui R., Li G., Gao Q., Yuan S., Altmeyer R., and Zou G. (2016) Teicoplanin inhibits Ebola pseudovirus infection in cell culture. Antiviral Res. 125, 1–7
    1. Zufferey R., Nagy D., Mandel R. J., Naldini L., and Trono D. (1997) Multiply attenuated lentiviral vector achieves efficient gene delivery in vivo. Nat. Biotechnol. 15, 871–875
    1. Watt A., Moukambi F., Banadyga L., Groseth A., Callison J., Herwig A., Ebihara H., Feldmann H., and Hoenen T. (2014) A novel life cycle modeling system for Ebola virus shows a genome length-dependent role of VP24 in virus infectivity. J. Virol. 88, 10511–10524
    1. Pols M. S., ten Brink C., Gosavi P., Oorschot V., and Klumperman J. (2013) The HOPS proteins hVps41 and hVps39 are required for homotypic and heterotypic late endosome fusion. Traffic 14, 219–232
    1. Ebert D. H., Deussing J., Peters C., and Dermody T. S. (2002) Cathepsin L and cathepsin B mediate reovirus disassembly in murine fibroblast cells. J. Biol. Chem. 277, 24609–24617
    1. Simmons G., Gosalia D. N., Rennekamp A. J., Reeves J. D., Diamond S. L., and Bates P. (2005) Inhibitors of cathepsin L prevent severe acute respiratory syndrome coronavirus entry. Proc. Natl. Acad. Sci. U.S.A. 102, 11876–11881
    1. Ryabchikova E. I., Kolesnikova L. V., and Luchko S. V. (1999) An analysis of features of pathogenesis in two animal models of Ebola virus infection. J. Infect. Dis. 179, S199–S202
    1. Geisbert T. W., Young H. A., Jahrling P. B., Davis K. J., Larsen T., Kagan E., and Hensley L. E. (2003) Pathogenesis of Ebola hemorrhagic fever in primate models: evidence that hemorrhage is not a direct effect of virus-induced cytolysis of endothelial cells. Am. J. Pathol. 163, 2371–2382
    1. Côté M., Misasi J., Ren T., Bruchez A., Lee K., Filone C. M., Hensley L., Li Q., Ory D., Chandran K., and Cunningham J. (2011) Small molecule inhibitors reveal Niemann-Pick C1 is essential for Ebola virus infection. Nature 477, 344–348
    1. Li W., Moore M. J., Vasilieva N., Sui J., Wong S. K., Berne M. A., Somasundaran M., Sullivan J. L., Luzuriaga K., Greenough T. C., Choe H., and Farzan M. (2003) Angiotensin-converting enzyme 2 is a functional receptor for the SARS coronavirus. Nature 426, 450–454
    1. Bröcker C., Kuhlee A., Gatsogiannis C., Balderhaar H. J., Hönscher C., Engelbrecht-Vandré S., Ungermann C., and Raunser S. (2012) Molecular architecture of the multisubunit homotypic fusion and vacuole protein sorting (HOPS) tethering complex. Proc. Natl. Acad. Sci. U.S.A. 109, 1991–1996
    1. Seals D. F., Eitzen G., Margolis N., Wickner W. T., and Price A. (2000) A Ypt/Rab effector complex containing the Sec1 homolog Vps33p is required for homotypic vacuole fusion. Proc. Natl. Acad. Sci. U.S.A. 97, 9402–9407
    1. Shirato K., Kawase M., and Matsuyama S. (2013) Middle East respiratory syndrome coronavirus infection mediated by the transmembrane serine protease TMPRSS2. J. Virol. 87, 12552–12561
    1. Parenti F., Beretta G., Berti M., and Arioli V. (1978) Teichomycins, new antibiotics from Actinoplanes teichomyceticus Nov. Sp. I. Description of the producer strain, fermentation studies and biological properties. J. Antibiot. 31, 276–283
    1. Borghi A., Coronelli C., Faniuolo L., Allievi G., Pallanza R., and Gallo G. G. (1984) Teichomycins, new antibiotics from Actinoplanes teichomyceticus nov. sp. IV. Separation and characterization of the components of teichomycin (teicoplanin). J. Antibiot. 37, 615–620
    1. Corti A., Soffientini A., and Cassani G. (1985) Binding of the glycopeptide antibiotic teicoplanin to d-alanyl-d-alanine-agarose: the effect of micellar aggregates. J. Appl. Biochem. 7, 133–137
    1. Corry J., Johnson S. M., Cornwell J., and Peeples M. E. (2015) Preventing cleavage of the respiratory syncytial virus attachment protein in Vero cells rescues the infectivity of progeny virus for primary human airway cultures. J. Virol. 10.1128/JVI.02351-15
    1. Balzarini J., Pannecouque C., De Clercq E., Pavlov A. Y., Printsevskaya S. S., Miroshnikova O. V., Reznikova M. I., and Preobrazhenskaya M. N. (2003) Antiretroviral activity of semisynthetic derivatives of glycopeptide antibiotics. J. Med. Chem. 46, 2755–2764
    1. Preobrazhenskaya M. N., and Olsufyeva E. N. (2006) Polycyclic peptide and glycopeptide antibiotics and their derivatives as inhibitors of HIV entry. Antiviral Res. 71, 227–236
    1. Bereczki I., Kicsák M., Dobray L., Borbás A., Batta G., Kéki S., Nikodém É. N., Ostorházi E., Rozgonyi F., Vanderlinden E., Naesens L., and Herczegh P. (2014) Semisynthetic teicoplanin derivatives as new influenza virus binding inhibitors: synthesis and antiviral studies. Bioorg. Med. Chem. Lett. 24, 3251–3254
    1. Maieron A., and Kerschner H. (2012) Teicoplanin therapy leading to a significant decrease in viral load in a patient with chronic hepatitis C. J. Antimicrob. Chemother. 67, 2537–2538
    1. Obeid S., Printsevskaya S. S., Olsufyeva E. N., Dallmeier K., Durantel D., Zoulim F., Preobrazhenskaya M. N., Neyts J., and Paeshuyse J. (2011) Inhibition of hepatitis C virus replication by semi-synthetic derivatives of glycopeptide antibiotics. J. Antimicrob. Chemother. 66, 1287–1294
    1. De Burghgraeve T., Kaptein S. J., Ayala-Nunez N. V., Mondotte J. A., Pastorino B., Printsevskaya S. S., de Lamballerie X., Jacobs M., Preobrazhenskaya M., Gamarnik A. V., Smit J. M., and Neyts J. (2012) An analog of the antibiotic teicoplanin prevents flavivirus entry in vitro. PLoS ONE 7, e37244.
    1. Balzarini J., Keyaerts E., Vijgen L., Egberink H., De Clercq E., Van Ranst M., Printsevskaya S. S., Olsufyeva E. N., Solovieva S. E., and Preobrazhenskaya M. N. (2006) Inhibition of feline (FIPV) and human (SARS) coronavirus by semisynthetic derivatives of glycopeptide antibiotics. Antiviral Res. 72, 20–33
    1. Andrault P. M., Samsonov S. A., Weber G., Coquet L., Nazmi K., Bolscher J. G., Lalmanach A. C., Jouenne T., Brömme D., Pisabarro M. T., Lalmanach G., and Lecaille F. (2015) Antimicrobial peptide LL-37 is both a substrate of cathepsins S and K and a selective inhibitor of cathepsin L. Biochemistry 54, 2785–2798
    1. Brogden R. N., and Peters D. H. (1994) Teicoplanin. A reappraisal of its antimicrobial activity, pharmacokinetic properties and therapeutic efficacy. Drugs 47, 823–854
    1. Cavalcanti A. B., Goncalves A. R., Almeida C. S., Bugano D. D., and Silva E. (2010) Teicoplanin versus vancomycin for proven or suspected infection. Cochrane Database Syst. Rev. 16, CD007022

Source: PubMed

3
Abonnere