Efficacy and Mechanism of Action of Low Dose Emetine against Human Cytomegalovirus

Rupkatha Mukhopadhyay, Sujayita Roy, Rajkumar Venkatadri, Yu-Pin Su, Wenjuan Ye, Elena Barnaeva, Lesley Mathews Griner, Noel Southall, Xin Hu, Amy Q Wang, Xin Xu, Andrés E Dulcey, Juan J Marugan, Marc Ferrer, Ravit Arav-Boger, Rupkatha Mukhopadhyay, Sujayita Roy, Rajkumar Venkatadri, Yu-Pin Su, Wenjuan Ye, Elena Barnaeva, Lesley Mathews Griner, Noel Southall, Xin Hu, Amy Q Wang, Xin Xu, Andrés E Dulcey, Juan J Marugan, Marc Ferrer, Ravit Arav-Boger

Abstract

Infection with human cytomegalovirus (HCMV) is a threat for pregnant women and immunocompromised hosts. Although limited drugs are available, development of new agents against HCMV is desired. Through screening of the LOPAC library, we identified emetine as HCMV inhibitor. Additional studies confirmed its anti-HCMV activities in human foreskin fibroblasts: EC50-40±1.72 nM, CC50-8±0.56 μM, and selectivity index of 200. HCMV inhibition occurred after virus entry, but before DNA replication, and resulted in decreased expression of viral proteins. Synergistic virus inhibition was achieved when emetine was combined with ganciclovir. In a mouse CMV (MCMV) model, emetine was well-tolerated, displayed long half-life, preferential distribution to tissues over plasma, and effectively suppressed MCMV. Since the in vitro anti-HCMV activity of emetine decreased significantly in low-density cells, a mechanism involving cell cycle regulation was suspected. HCMV inhibition by emetine depended on ribosomal processing S14 (RPS14) binding to MDM2, leading to disruption of HCMV-induced MDM2-p53 and MDM2-IE2 interactions. Irrespective of cell density, emetine induced RPS14 translocation into the nucleus during infection. In infected high-density cells, MDM2 was available for interaction with RPS14, resulting in disruption of MDM2-p53 interaction. However, in low-density cells the pre-existing interaction of MDM2-p53 could not be disrupted, and RPS14 could not interact with MDM2. In high-density cells the interaction of MDM2-RPS14 resulted in ubiquitination and degradation of RPS14, which was not observed in low-density cells. In infected-only or in non-infected emetine-treated cells, RPS14 failed to translocate into the nucleus, hence could not interact with MDM2, and was not ubiquitinated. HCMV replicated similarly in RPS14 knockdown or control cells, but emetine did not inhibit virus replication in the former cell line. The interaction of MDM2-p53 was maintained in infected RPS14 knockdown cells despite emetine treatment, confirming a unique mechanism by which emetine exploits RPS14 to disrupt MDM2-p53 interaction. Summarized, emetine may represent a promising candidate for HCMV therapy alone or in combination with ganciclovir through a novel host-dependent mechanism.

Conflict of interest statement

The authors have declared that no competing interests exist.

Figures

Fig 1. Anti-HCMV activity of emetine and…
Fig 1. Anti-HCMV activity of emetine and synergy with GCV.
A) Cells were infected with pp28-luciferase HCMV Towne and treated with indicated concentrations of emetine or GCV. Luciferase activity was measured at 72 hpi. Data represent mean ± SE of triplicate determinations from a representative of three independent experiments. B) Cells were treated with the indicated concentrations of emetine and cell viability was determined after 72 h. Data represent mean values ± SE of triplicate determinations from a representative of three independent experiments. C) Inhibition of HCMV protein expression by emetine (75 nM) or GCV (5 μM) at 72 hpi. Cells were infected with HCMV Towne and treated with emetine. Western blot of viral proteins IE1/2, UL44 and pp65 was performed at 72 hpi. D) Cells were infected with pp28-luciferase HCMV Towne, and treated with combination of GCV and emetine. The antiviral activity was evaluated by luciferase assay. Data represent mean values ± the SE of triplicate determinations from a representative of three independent experiments.
Fig 2. Emetine is an early inhibitor…
Fig 2. Emetine is an early inhibitor of HCMV replication.
A) Emetine does not inhibit HCMV entry. Cells were treated with emetine (75 nM), GCV (10 μM), and CpG 2006 (10 μM) 24 h prior to infection. Cells were infected with HCMV and treated with compounds for 90 min. Immunofluorescence staining was performed with mouse monoclonal anti-pp65 antibody. The fluorescence of rhodamine anti-mouse IgG and DAPI was visualized and merged using a Nikon Eclipse E-800 fluorescence microscope. B) Emetine has an early activity against HCMV. Cells were infected with HCMV Towne, and compounds were added at 0, 6, 12, 24, 36, and 48 hpi (Add on). Culture supernatants (10%) were collected at 72 hpi for a plaque assay after 14 days. C) Cells were infected with HCMV Towne and treated with compounds immediately after virus adsorption. Compounds were removed at 0, 6, 12, 24, 36, and 48 hpi (Removal). Culture supernatants were collected at 72 hpi for titration by plaque assay. Data represent mean ± SE of triplicate determinations from a representative of two independent experiments.
Fig 3. Emetine achieves high tissue concentrations…
Fig 3. Emetine achieves high tissue concentrations and is efficacious against MCMV replication.
A)In vivo pharmacokinetics of emetine. Plasma, liver, lung and spleen samples were collected at the indicated time points after single oral administration of emetine at 0.1 mg/kg in male BALB/c mice. Concentrations were measured using UPLC-MS/MS methods. For plasma, the concentrations observed were mostly below the level limit of quantification. B) Quantitative real-time PCR of viral gB was performed on DNA extracted from blood at day 14 post infection. Plaque assay was performed from salivary glands C) or liver D) collected at day 14 post infection. Emetine was administered orally starting 24 hpi or 72 hpi at 0.1 or 1.0 mg/kg every 3 days. GCV dose was 10 mg/kg/dose administered intraperitoneally twice daily.
Fig 4. The anti-HCMV activity of emetine…
Fig 4. The anti-HCMV activity of emetine is reduced in low-density HFFs.
A) Cells were seeded at indicated densities in a 96-well plate, infected with pp28-luciferase HCMV Towne (MOI = 1) and treated with emetine (75 nM) or GCV (5 μM). Luciferase activity was measured at 72 hpi and normalized to the activity of infected untreated cells. Data represent mean ± SE of triplicate determinations from a representative of two independent experiments. B) Same cell lysates as in A were collected for Western blotting at 72 hpi and expression of viral proteins UL44 and pp65 was determined. C) Supernatants from each well as in A were used to infect cells (1 million cells in a 96-well plate) in the corresponding well and luciferase activity was measured at 72 hpi. D) Cells were seeded into 12-well plates at 0.5 and 2 million cells/plate, infected with 100 PFU/well of HCMV Towne or MCMV, and treated with emetine (75 nM) or GCV (5 μM). After 10 days (for HCMV) or 3 days (for MCMV) plaques were stained with crystal violet and the number of plaques enumerated. Data shown are average of 2 wells (±SD) for a representative experiment from two different experiments.
Fig 5. Emetine induces an interaction between…
Fig 5. Emetine induces an interaction between MDM2 and RPS14 in infected high-density cells.
A) Emetine abrogates infection-mediated overexpression of RPS14 at 72 hpi. Cells were seeded at 0.5 or 2 million cells/plate, infected with Towne HCMV and treated with emetine (75 nM) or GCV (5μM). Lysates were collected at 24 or 72 hpi for Western blotting. B) HFFs were seeded at 1 or 2 million cells/plate and treated with emetine or GCV. Lysates were collected at 72 hpi for Western blotting. C) Cells were seeded at 0.5 or 2 million/plate in 100 mm dishes, infected with Towne HCMV followed by treatment with emetine (75 nM) or GCV (5μM) for 24h. MG132 (10 μM) was added after 12 h. At 24 hpi, lysates were collected and subjected to IP with anti-MDM2 followed by immunoblotting with anti-RPS14 antibody or D) In reverse reaction, IP was performed with anti-RPS14 followed by immunoblotting with anti-MDM2 antibody. Densitometry analysis was performed to normalize immunoprecipitated protein to its input level. E) A model showing the interaction of MDM2 and RPS14 in high-density but not in low-density cells.
Fig 6. Emetine induces nuclear translocation of…
Fig 6. Emetine induces nuclear translocation of RPS14 in infected high-density cells at 24 hpi followed by cytoplasmic relocalization.
A) Cells were seeded at 2 million/4-well chamber slide, infected and treated with either emetine (75 nM) or GCV (5μM) for 24 h. Cells were stained with IE1/2 (Alexa 555:Red) for evidence of infection and RPS14 (FITC: Green) and nuclear DAPI. Stained slides were subjected to confocal microscopy and colocalization was studied and quantified using NIS elements software (Nikon). B) The same procedure was repeated at 72 hpi. The images shown are a representative of two independent experiments. Percent nuclear localization is represented as Mean ± SD from two different fields of at least 40 cells per condition.
Fig 7. Nuclear localization of RPS14 persists…
Fig 7. Nuclear localization of RPS14 persists during infection and emetine treatment in low-density cells.
A) Cells were seeded at 0.5 million/4-well chamber slide, infected and treated with either emetine (75 nM) or GCV (5μM) for 24 h. Cells were stained with IE1/2 (Alexa 555:Red) for evidence of infection and RPS14 (FITC: Green) and nuclear DAPI. Stained slides were subjected to confocal microscopy and nuclear localization was quantified using NIS elements software (Nikon). B) The same procedure was repeated at 72 hpi. The images shown are a representative of two independent experiments. Percent nuclear localization is represented as mean ± SD of at least 25 cells per condition from two different fields in the slide.
Fig 8. RPS14 is ubiquitinated and degraded…
Fig 8. RPS14 is ubiquitinated and degraded in infected high-density cells.
A) Cells were seeded at 0.5 or 2 million/plate in 100 mm dishes, infected with HCMV Towne followed by treatment with emetine (75 nM) or GCV (5μM) for 72h. MG132 (10 μM) was added 12 h before harvest. Lysates were collected at 72 hpi and subjected to IP with anti-Ubiquitin antibody followed by immunoblotting with anti-RPS14 antibody or IP with anti-RPS14 antibody followed by immunoblotting with anti-Ubiquitin antibody. The mono- and poly- ubiquitinated forms of RPS14 are depicted as (Ub)-RPS14 and n(Ub)-RPS14, respectively. B) HCMV-infected cells (left: high-density right: low-density) were treated with emetine or GCV. At 24 hpi cycloheximide (CHX, 100 μg/mL) was added. Cells were harvested at the indicated time intervals up to 8 h following CHX for SDS-PAGE analysis.
Fig 9. Emetine disrupts HCMV mediated MDM2-p53…
Fig 9. Emetine disrupts HCMV mediated MDM2-p53 complex.
A) MDM2 and p53 expression increases with emetine treatment. Cells were seeded at 0.5 or 2 million cells/plate, infected with HCMV Towne followed by treatment with emetine (75 nM) or GCV (5 μM). Cell lysates were harvested at 24 or 72 hpi for Western blotting. B) Cells were seeded at 2 million cells/plate, infected with HCMV Towne and treated with emetine (75 nM) or GCV (5 μM). RNA was harvested and qRT-PCR was performed for p21. Data represent mean ±SE of triplicate determinations from a representative of three independent experiments C) HFFs were seeded at 1 or 2 million cells/plate and treated with emetine (75 nM) or GCV (5 μM). Cell lysates were harvested at 72 hpi for Western blotting. D) HFFs were seeded at either 0.5 or 2 million/plate in 100 mm dishes, infected with HCMV Towne followed by treatment with emetine (75 nM) or GCV (5μM) for 24h. MG132 (10 μM) was added after 12 h. At 24 hpi, lysates were collected and subjected to IP with anti-MDM2 antibody followed by immunoblotting with anti-p53 antibody or E) IP with anti-p53 antibody followed by immunoblotting with anti-MDM2 antibody. F) A model depicting the mechanism by which emetine disrupts HCMV mediated MDM2-p53 interaction in high-density cells, but not in cycling low-density cells.
Fig 10. Emetine loses its anti-HCMV activity…
Fig 10. Emetine loses its anti-HCMV activity in RPS14 knockdown cells.
A) RPS14 knockdown HFFs (sh-RPS14) were generated from lentiviral system and expression of RPS14 in the knockdown cells was compared to TRC control cells by Western blotting. B) sh-RPS14 and TRC control cells were seeded at 2 million/plate, infected and treated with emetine (75 nM) or GCV (5μM) for 72h. Cell viability was determined after 72 h. C) Luciferase activity was measured in cell lysates. D) sh-RPS14 and TRC control cells were seeded at 2 million/plate, in a 12-well plate and infected 24 hours later at 100 PFU/well. After 90 min virus was aspirated, and DMEM containing 4% fetal bovine serum (FBS) with 0.5% carboxymethyl-cellulose, were added with the compounds at indicated concentrations into triplicate wells. Plaques were counted after incubation at 37°C for 8 days. E) Same cell lysates as in B were used to determine HCMV pp65, UL44 and IE1/2 expression. F) sh-RPS14 and TRC-control cells were infected and treated with emetine (75 nM) for 24h. MG132 (10 μM) was added after 12 h. At 24 hpi, lysates were collected and subjected to IP with anti-p53 antibody followed by immunoblotting with anti-MDM2 antibody.
Fig 11. Model of HCMV inhibition by…
Fig 11. Model of HCMV inhibition by emetine.
In high-density infected cells (A) emetine induces (1) nuclear translocation of RPS14 (2) followed by RPS14 binding to MDM2 (3 & 4) resulting in disruption of the interaction between MDM2-p53 (6) and MDM2- viral IE2 (5 & 7), and by RPS14 ubiquitination and degradation (8). In low-density infected cells (B) although emetine induces (1) nuclear translocation of RPS14 (2), it is unable to interact with MDM2 (4) which is already bound to p53 to facilitate virus replication (3).

References

    1. Griffiths PD, Clark DA, Emery VC (2000) Betaherpesviruses in transplant recipients. J Antimicrob Chemother 45 Suppl T3: 29–34.
    1. Kovacs A, Schluchter M, Easley K, Demmler G, Shearer W et al. (1999) Cytomegalovirus infection and HIV-1 disease progression in infants born to HIV-1-infected women. Pediatric Pulmonary and Cardiovascular Complications of Vertically Transmitted HIV Infection Study Group. N Engl J Med 341: 77–84.
    1. Demmler GJ (1991) Infectious Diseases Society of America and Centers for Disease Control. Summary of a workshop on surveillance for congenital cytomegalovirus disease. Rev Infect Dis 13: 315–329.
    1. Manicklal S, Emery VC, Lazzarotto T, Boppana SB, Gupta RK (2013) The "silent" global burden of congenital cytomegalovirus. Clin Microbiol Rev 26: 86–102. 26/1/86 [pii];10.1128/CMR.00062-12
    1. Prichard MN, Kern ER (2011) The search for new therapies for human cytomegalovirus infections. Virus Res 157: 212–221. 10.1016/j.virusres.2010.11.004
    1. Schreiber A, Harter G, Schubert A, Bunjes D, Mertens T et al. (2009) Antiviral treatment of cytomegalovirus infection and resistant strains. Expert Opin Pharmacother 10: 191–209. 10.1517/14656560802678138
    1. Steininger C (2007) Novel therapies for cytomegalovirus disease. Recent Pat Antiinfect Drug Discov 2: 53–72.
    1. Kimberlin DW, Lin CY, Sanchez PJ, Demmler GJ, Dankner W et al. (2003) Effect of ganciclovir therapy on hearing in symptomatic congenital cytomegalovirus disease involving the central nervous system: a randomized, controlled trial. J Pediatr 143: 16–25.
    1. Kimberlin DW, Jester PM, Sanchez PJ, Ahmed A, Arav-Boger R et al. (2015) Valganciclovir for symptomatic congenital cytomegalovirus disease. N Engl J Med 372: 933–943. 10.1056/NEJMoa1404599
    1. Jabs DA, Martin BK, Forman MS (2010) Mortality associated with resistant cytomegalovirus among patients with cytomegalovirus retinitis and AIDS. Ophthalmology 117: 128–132. 10.1016/j.ophtha.2009.06.016
    1. Chou SW (2001) Cytomegalovirus drug resistance and clinical implications. Transpl Infect Dis 3 Suppl 2: 20–24.
    1. Krosky PM, Baek MC, Coen DM (2003) The human cytomegalovirus UL97 protein kinase, an antiviral drug target, is required at the stage of nuclear egress. J Virol 77: 905–914.
    1. Williams SL, Hartline CB, Kushner NL, Harden EA, Bidanset DJ et al. (2003) In vitro activities of benzimidazole D- and L-ribonucleosides against herpesviruses. Antimicrob Agents Chemother 47: 2186–2192.
    1. Lischka P, Hewlett G, Wunberg T, Baumeister J, Paulsen D et al. (2010) In vitro and in vivo activities of the novel anticytomegalovirus compound AIC246. Antimicrob Agents Chemother 54: 1290–1297. 10.1128/AAC.01596-09
    1. Kaul DR, Stoelben S, Cober E, Ojo T, Sandusky E et al. (2011) First report of successful treatment of multidrug-resistant cytomegalovirus disease with the novel anti-CMV compound AIC246. Am J Transplant 11: 1079–1084. 10.1111/j.1600-6143.2011.03530.x
    1. Winston DJ, Young JA, Pullarkat V, Papanicolaou GA, Vij R et al. (2008) Maribavir prophylaxis for prevention of cytomegalovirus infection in allogeneic stem cell transplant recipients: a multicenter, randomized, double-blind, placebo-controlled, dose-ranging study. Blood 111: 5403–5410. 10.1182/blood-2007-11-121558
    1. Shen L, Peterson S, Sedaghat AR, McMahon MA, Callender M et al. (2008) Dose-response curve slope sets class-specific limits on inhibitory potential of anti-HIV drugs. Nat Med 14: 762–766. 10.1038/nm1777
    1. Thoene JG, Lemons R, Boskovich S, Borysko K (1985) Inhibitors of protein synthesis also inhibit lysosomal proteolysis. Studies using cystinotic fibroblasts. J Clin Invest 75: 370–376.
    1. Entner N, Grollman AP (1973) Inhibition of protein synthesis: a mechanism of amebicide action of emetine and other structurally related compounds. J Protozool 20: 160–163.
    1. Madjar JJ, Nielsen-Smith K, Frahm M, Roufa DJ (1982) Emetine resistance in chinese hamster ovary cells is associated with an altered ribosomal protein S14 mRNA. Proc Natl Acad Sci U S A 79: 1003–1007.
    1. Zhou X, Hao Q, Liao J, Zhang Q, Lu H (2013) Ribosomal protein S14 unties the MDM2-p53 loop upon ribosomal stress. Oncogene 32: 388–396. 10.1038/onc.2012.63
    1. Riley MF, Lozano G (2012) The Many Faces of MDM2 Binding Partners. Genes Cancer 3: 226–239. 10.1177/1947601912455322
    1. Kulikov R, Winter M, Blattner C (2006) Binding of p53 to the central domain of Mdm2 is regulated by phosphorylation. J Biol Chem 281: 28575–28583.
    1. Moll UM, Petrenko O (2003) The MDM2-p53 interaction. Mol Cancer Res 1: 1001–1008.
    1. Worrall EG, Wawrzynow B, Worrall L, Walkinshaw M, Ball KL et al. (2009) Regulation of the E3 ubiquitin ligase activity of MDM2 by an N-terminal pseudo-substrate motif. J Chem Biol 2: 113–129. 10.1007/s12154-009-0019-5
    1. Zhang Z, Evers DL, McCarville JF, Dantonel JC, Huong SM et al. (2006) Evidence that the human cytomegalovirus IE2-86 protein binds mdm2 and facilitates mdm2 degradation. J Virol 80: 3833–3843. 80/8/3833 [pii];10.1128/JVI.80.8.3833-3843.2006
    1. Lurain NS, Chou S (2010) Antiviral drug resistance of human cytomegalovirus. Clin Microbiol Rev 23: 689–712. 10.1128/CMR.00009-10
    1. Roy S, Arav-Boger R (2014) New cell-signaling pathways for controlling cytomegalovirus replication. Am J Transplant 14: 1249–1258. 10.1111/ajt.12725
    1. Kern ER, Collins DJ, Wan WB, Beadle JR, Hostetler KY et al. (2004) Oral treatment of murine cytomegalovirus infections with ether lipid esters of cidofovir. Antimicrob Agents Chemother 48: 3516–3522.
    1. Madjar JJ, Frahm M, McGill S, Roufa DJ (1983) Ribosomal protein S14 is altered by two-step emetine resistance mutations in Chinese hamster cells. Mol Cell Biol 3: 190–197.
    1. Browne EP, Wing B, Coleman D, Shenk T (2001) Altered cellular mRNA levels in human cytomegalovirus-infected fibroblasts: viral block to the accumulation of antiviral mRNAs. J Virol 75: 12319–12330.
    1. Hwang ES, Zhang Z, Cai H, Huang DY, Huong SM et al. (2009) Human cytomegalovirus IE1-72 protein interacts with p53 and inhibits p53-dependent transactivation by a mechanism different from that of IE2-86 protein. J Virol 83: 12388–12398. JVI.00304-09 [pii];10.1128/JVI.00304-09
    1. Casavant NC, Luo MH, Rosenke K, Winegardner T, Zurawska A et al. (2006) Potential role for p53 in the permissive life cycle of human cytomegalovirus. J Virol 80: 8390–8401. 80/17/8390 [pii];10.1128/JVI.00505-06
    1. Fortunato EA, Spector DH (1998) p53 and RPA are sequestered in viral replication centers in the nuclei of cells infected with human cytomegalovirus. J Virol 72: 2033–2039.
    1. Chen Z, Knutson E, Wang S, Martinez LA, Albrecht T (2007) Stabilization of p53 in human cytomegalovirus-initiated cells is associated with sequestration of HDM2 and decreased p53 ubiquitination. J Biol Chem 282: 29284–29295. M705349200 [pii];10.1074/jbc.M705349200
    1. Haupt Y, Maya R, Kazaz A, Oren M (1997) Mdm2 promotes the rapid degradation of p53. Nature 387: 296–299.
    1. Kubbutat MH, Jones SN, Vousden KH (1997) Regulation of p53 stability by Mdm2. Nature 387: 299–303.
    1. Diaz JJ, Roufa DJ (1992) Fine-structure map of the human ribosomal protein gene RPS14. Mol Cell Biol 12: 1680–1686.
    1. Boultwood J (2011) The role of haploinsufficiency of RPS14 and p53 activation in the molecular pathogenesis of the 5q- syndrome. Pediatr Rep 3 Suppl 2: e10 10.4081/pr.2011.s2.e10 pr.2011.s2.e10 [pii].
    1. Panettiere F, Coltman CA Jr. (1971) Experience with emetine hydrochloride (NSC 33669) as an antitumor agent. Cancer 27: 835–841.
    1. Mastrangelo MJ, Grage TB, Bellet RE, Weiss AJ (1973) A phase I study of emetine hydrochloride (NSC 33669) in solid tumors. Cancer 31: 1170–1175.
    1. Reagan-Shaw S, Nihal M, Ahmad N (2008) Dose translation from animal to human studies revisited. FASEB J 22: 659–661. fj.07-9574LSF [pii];10.1096/fj.07-9574LSF
    1. Regenthal R, Krueger M, Koeppel C, Preiss R (1999) Drug levels: therapeutic and toxic serum/plasma concentrations of common drugs. J Clin Monit Comput 15: 529–544.
    1. Knight R (1980) The chemotherapy of amoebiasis. J Antimicrob Chemother 6: 577–593.
    1. Muhammad I, Dunbar DC, Khan SI, Tekwani BL, Bedir E et al. (2003) Antiparasitic alkaloids from Psychotria klugii. J Nat Prod 66: 962–967.
    1. Carney RW, Wojtkunski J, Konopka EA, deStevens G (1966) The chemical, spectral, and biological properties of monomethine cyanine dyes containing 1,3-benzoxazine and quinazoline nuclei. J Med Chem 9: 758–762.
    1. Rosenkranz V, Wink M (2008) Alkaloids induce programmed cell death in bloodstream forms of trypanosomes (Trypanosoma b. brucei). Molecules 13: 2462–2473. 10.3390/molecules13102462
    1. Deng L, Dai P, Ciro A, Smee DF, Djaballah H et al. (2007) Identification of novel antipoxviral agents: mitoxantrone inhibits vaccinia virus replication by blocking virion assembly. J Virol 81: 13392–13402.
    1. Chaves Valadao AL, Abreu CM, Dias JZ, Arantes P, Verli H et al. (2015) Natural Plant Alkaloid (Emetine) Inhibits HIV-1 Replication by Interfering with Reverse Transcriptase Activity. Molecules 20: 11474–11489. molecules200611474 [pii];10.3390/molecules200611474
    1. He R, Sandford G, Hayward GS, Burns WH, Posner GH et al. (2011) Recombinant Luciferase-Expressing Human Cytomegalovirus (CMV) for evaluation of CMV inhibitors. Virol J 8: 40 10.1186/1743-422X-8-40
    1. Lurain NS, Chou S (2010) Antiviral drug resistance of human cytomegalovirus. Clin Microbiol Rev 23: 689–712. 10.1128/CMR.00009-10
    1. Summers BC, Margolis TP, Leib DA (2001) Herpes simplex virus type 1 corneal infection results in periocular disease by zosteriform spread. J Virol 75: 5069–5075.
    1. Fischer L, Laib SK, Jahn G, Hamprecht K, Gohring K (2013) Generation and characterization of a GCV resistant HCMV UL97-mutation and a drug sensitive UL54-mutation. Antiviral Res 100: 575–577. S0166-3542(13)00286-6 [pii];10.1016/j.antiviral.2013.09.026
    1. Cai H, Kapoor A, He R, Venkatadri R, Forman M et al. (2014) In vitro combination of anti-cytomegalovirus compounds acting through different targets: role of the slope parameter and insights into mechanisms of Action. Antimicrob Agents Chemother 58: 986–994. AAC.01972-13 [pii];10.1128/AAC.01972-13
    1. Bliss C.I. (1939) The toxicity of poisons applied jointly. Ann Appl Biol 26: 585–615.
    1. Jilek BL, Zarr M, Sampah ME, Rabi SA, Bullen CK et al. (2012) A quantitative basis for antiretroviral therapy for HIV-1 infection. Nat Med 18: 446–451. 10.1038/nm.2649
    1. Liu J, Xu Y, Stoleru D, Salic A (2012) Imaging protein synthesis in cells and tissues with an alkyne analog of puromycin. Proc Natl Acad Sci U S A 109: 413–418. 1111561108 [pii];10.1073/pnas.1111561108
    1. Iversen AC, Steinkjer B, Nilsen N, Bohnhorst J, Moen SH et al. (2009) A proviral role for CpG in cytomegalovirus infection. J Immunol 182: 5672–5681. 10.4049/jimmunol.0801268
    1. Vliegen I, Herngreen S, Grauls G, Bruggeman C, Stassen F (2003) Improved detection and quantification of mouse cytomegalovirus by real-time PCR. Virus Res 98: 17–25.
    1. Lal A, Haynes SR, Gorospe M (2005) Clean Western blot signals from immunoprecipitated samples. Mol Cell Probes 19: 385–388. S0890-8508(05)00044-7 [pii];10.1016/j.mcp.2005.06.007
    1. Pizzorno MC, O'Hare P, Sha L, LaFemina RL, Hayward GS (1988) trans-activation and autoregulation of gene expression by the immediate-early region 2 gene products of human cytomegalovirus. J Virol 62: 1167–1179.
    1. Ebert BL, Pretz J, Bosco J, Chang CY, Tamayo P et al. (2008) Identification of RPS14 as a 5q- syndrome gene by RNA interference screen. Nature 451: 335–339. nature06494 [pii];10.1038/nature06494
    1. Caceres G, McGraw K, Yip BH, Pellagatti A, Johnson J et al. (2013) TP53 suppression promotes erythropoiesis in del(5q) MDS, suggesting a targeted therapeutic strategy in lenalidomide-resistant patients. Proc Natl Acad Sci U S A 110: 16127–16132. 1311055110 [pii];10.1073/pnas.1311055110
    1. Tiscornia G, Singer O, Ikawa M, Verma IM (2003) A general method for gene knockdown in mice by using lentiviral vectors expressing small interfering RNA. Proc Natl Acad Sci U S A 100: 1844–1848.
    1. Kapoor A, Forman M, Arav-Boger R (2014) Activation of nucleotide oligomerization domain 2 (NOD2) by human cytomegalovirus initiates innate immune responses and restricts virus replication. PLoS One 9: e92704 10.1371/journal.pone.0092704 PONE-D-13-49464 [pii].

Source: PubMed

3
購読する