MERS-CoV pathogenesis and antiviral efficacy of licensed drugs in human monocyte-derived antigen-presenting cells

Yu Cong, Brit J Hart, Robin Gross, Huanying Zhou, Matthew Frieman, Laura Bollinger, Jiro Wada, Lisa E Hensley, Peter B Jahrling, Julie Dyall, Michael R Holbrook, Yu Cong, Brit J Hart, Robin Gross, Huanying Zhou, Matthew Frieman, Laura Bollinger, Jiro Wada, Lisa E Hensley, Peter B Jahrling, Julie Dyall, Michael R Holbrook

Abstract

Middle East respiratory syndrome coronavirus (MERS-CoV) presents an emerging threat to public health worldwide by causing severe respiratory disease in humans with high virulence and case fatality rate (about 35%) since 2012. Little is known about the pathogenesis and innate antiviral response in primary human monocyte-derived macrophages (MDMs) and dendritic cells (MDDCs) upon MERS-CoV infection. In this study, we assessed MERS-CoV replication as well as induction of inflammatory cytokines and chemokines in MDMs and immature and mature MDDCs. Immature MDDCs and MDMs were permissive for MERS-CoV infection, while mature MDDCs were not, with stimulation of proinflammatory cytokine and chemokine upregulation in MDMs, but not in MDDCs. To further evaluate the antiviral activity of well-defined drugs in primary antigen presenting cells (APCs), three compounds (chloroquine, chlorpromazine and toremifine), each with broad-spectrum antiviral activity in immortalized cell lines, were evaluated in MDMs and MDDCs to determine their antiviral effect on MERS-CoV infection. While chloroquine was not active in these primary cells, chlorpromazine showed strong anti-MERS-CoV activity, but it was associated with high cytotoxicity narrowing the potential window for drug utilization. Unlike in established cells, toremifene had marginal activity when tested in antigen presenting cells, with high apparent cytotoxicity, also limiting its potential as a therapeutic option. These results demonstrate the value of testing drugs in primary cells, in addition to established cell lines, before initiating preclinical or clinical studies for MERS treatment and the importance of carefully assessing cytotoxicity in drug screen assays. Furthermore, these studies also highlight the role of APCs in stimulating a robust protective immune response to MERS-CoV infection.

Conflict of interest statement

Competing Interests: The authors have declared that they have no competing interests.

Figures

Fig 1. Differentiation of macrophages and dendritic…
Fig 1. Differentiation of macrophages and dendritic cells from peripheral blood mononuclear cells.
CD14+ monocytes were isolated from human peripheral blood mononuclear cells (PBMCs). Cells were then differentiated to monocyte-derived macrophages (MDMs) by adding macrophage-colony stimulating factor (M-CSF) or to immature monocyte-derived dendritic cells (MDDCs) by adding granulocyte macrophage-colony stimulating factor (GM-CSF) and interleukin-4 (IL-4). Immature MDDCs were exposed to MDDC maturation cocktail (R-10 medium containing 10 ng/ml of tumor necrosis factor (TNF)-α, 10 ng/ml of IL-1β, 15 ng/ml of IL-6, 1 μg/ml of prostaglandin E2 (PEG2), 20 ng/ml of GM-CSF, 10 ng/ml of IL-4) overnight for maturation. MDMs and MDDCs were characterized by flow cytometry for expression of major cell-surface markers. Using these protocols, the MDMs are predominantly CD14+, CD11b+, HLA-DR+, CD169+, CD163+, and CD86+. The MDDCs are predominantly CD14-, CD11c+, HLA-DR+, CD80+, and CD86+. Mature MDDCs are CD83+, while immature MDDCs are CD83-.
Fig 2. Viral titers from supernatants of…
Fig 2. Viral titers from supernatants of MERS-CoV-infected macrophages and dendritic cells via plaque assay.
(A) Macrophages (MDMs), (B) immature monocyte-derived dendritic cells (MDDCs), and (C) mature MDDCs were infected at an MOI = 2 with MERS-CoV Jordan variant. Culture supernatants were collected at indicated times post-inoculation to detect viral titers. Each line represents cells from each individual donor. Titrations were performed in triplicate with data points representing the mean of the triplicate values.
Fig 3. Cytokine and chemokine responses to…
Fig 3. Cytokine and chemokine responses to MERS-CoV infection from certain innate immune primary cells.
(A) Human macrophages (MDMs) (B), immature monocyte-derived (MDDCs), and (C) mature MDDCs were inoculated with MERS-CoV Jordan variant at an MOI = 2 or subjected to mock infection. From cell culture supernatants, the fold change in immunoregulatory cytokines and chemokines relative to that of mock-infected cells incubated for the same period of time, were determined by multiplex assay. All data points represent triplicate biological replicates from four donors.
Fig 4. Antiviral activity of three compounds…
Fig 4. Antiviral activity of three compounds on MERS-CoV infection in MDMs.
(A) Effect of pretreatment of monocyte-derived macrophages (MDMs) with toremifene (TOMF), (B) chloroquine (CQ), and (C) chlorpromazine (CPZ) prior to MERS-CoV exposure on subsequent infection. The effect of these compounds on the cytotoxicity of macrophages without virus was also studied. MERS-CoV S protein was detected in infected cells using an immunofluorescence assay. The antigen was detected with a rabbit polyclonal antibody to S protein followed by staining with Alexa Fluor 594-conjugated goat anti-rabbit antibody. Percent inhibition of MERS-CoV infection is shown in blue, and percent cytotoxicity of the compounds without virus is shown in orange. Dotted gray line indicates half maximal effective concentrations (EC50) and 50% cytotoxicity concentrations (CC50). The selectivity index (SI) is defined as CC50÷EC50. Results are representative of 2 individual experiments with 3 replicates in each experiment (means ± standard error of the means [SEM]).
Fig 5. Antiviral activity of three compounds…
Fig 5. Antiviral activity of three compounds on MERS-CoV infection in MDMs and immature MDDCs.
(A) Monocyte-derived macrophages (MDMs), (B) immature dendritic cells (MDDCs) and (C) Vero E6 cells were pretreated with toremifene (TOMF), chloroquine (CQ), and chlorpromazine (CPZ) for 1 h followed by inoculation with MERS-CoV Jordan variant at an MOI = 0.1. Supernatants collected at 72 h post infection (hpi) for APCs and 48 hpi for Vero E6 cells were inoculated onto Vero E6 cells pre-seeded overnight in 96 well plates and incubated for 6 days. Wells with cytopathic effects were counted, and TCID50 was determined using the Reed-Muench method. Cytotoxicity was measured in parallel using luminescent cell viability assay at 48 h after compounds addition to the mock-infected MDMs and immature MDDCs. Antiviral activity is shown in gray and cytotoxicity is shown in orange. Results are representative of 2 individual experiments with 3 replicates in each (means ± standard error of the means [SEM]).

References

    1. Zaki AM, van Boheemen S, Bestebroer TM, Osterhaus AD, Fouchier RA. Isolation of a novel coronavirus from a man with pneumonia in Saudi Arabia. N Engl J Med. 2012;367(19):1814–20. Epub 2012/10/19. doi: .
    1. de Groot RJ, Baker SC, Baric RS, Brown CS, Drosten C, Enjuanes L, et al. Middle East respiratory syndrome coronavirus (MERS-CoV): announcement of the Coronavirus Study Group. Journal of virology. 2013;87(14):7790–2. Epub 2013/05/17. doi: ; PubMed Central PMCID: PMCPmc3700179.
    1. World Health Organization. Middle East respiratory syndrome coronavirus (MERS-CoV)—Saudi Arabia Disease outbreak news. Geneva, CH: World Health Organization; 2017. [cited 2017 11 September]. Available from: .
    1. Almario CV, Chey W, Kaung A, Whitman C, Fuller G, Reid M, et al. Computer-generated vs. physician-documented history of present illness (HPI): results of a blinded comparison. Am J Gastroenterol. 2015;110(1):170–9. doi: ; PubMed Central PMCID: PMC4289091.
    1. Haagmans BL, Al Dhahiry SH, Reusken CB, Raj VS, Galiano M, Myers R, et al. Middle East respiratory syndrome coronavirus in dromedary camels: an outbreak investigation. Lancet Infect Dis. 2014;14(2):140–5. doi: .
    1. World Health Organization. Middle East respiratory syndrome coronavirus (MERS-CoV) Fact sheet. Geneva, CH: World Health Organization; 2017. [updated 2017 May; cited 2017 11 September]. Available from: .
    1. Hui DS, Memish ZA, Zumla A. Severe acute respiratory syndrome vs. the Middle East respiratory syndrome. Current opinion in pulmonary medicine. 2014;20(3):233–41. Epub 2014/03/15. doi: .
    1. Senga M, Arabi YM, Fowler RA. Clinical spectrum of the Middle East respiratory syndrome coronavirus (MERS-CoV). J Infect Public Health. 2017;10(2):191–4. doi: .
    1. Poissy J, Goffard A, Parmentier-Decrucq E, Favory R, Kauv M, Kipnis E, et al. Kinetics and pattern of viral excretion in biological specimens of two MERS-CoV cases. Journal of clinical virology: the official publication of the Pan American Society for Clinical Virology. 2014;61(2):275–8. Epub 2014/07/31. doi: .
    1. Collins RA, Werling D, Duggan SE, Bland AP, Parsons KR, Howard CJ. Gammadelta T cells present antigen to CD4+ alphabeta T cells. Journal of leukocyte biology. 1998;63(6):707–14. Epub 1998/06/10. .
    1. Funk CJ, Wang J, Ito Y, Travanty EA, Voelker DR, Holmes KV, et al. Infection of human alveolar macrophages by human coronavirus strain 229E. The Journal of general virology. 2012;93(Pt 3):494–503. Epub 2011/11/18. doi: ; PubMed Central PMCID: PMCPmc3352353.
    1. Kopf M, Schneider C, Nobs SP. The development and function of lung-resident macrophages and dendritic cells. Nature immunology. 2015;16(1):36–44. Epub 2014/12/19. doi: .
    1. Dyall J, Coleman CM, Hart BJ, Venkataraman T, Holbrook MR, Kindrachuk J, et al. Repurposing of clinically developed drugs for treatment of Middle East respiratory syndrome coronavirus infection. Antimicrobial agents and chemotherapy. 2014;58(8):4885–93. Epub 2014/05/21. doi: ; PubMed Central PMCID: PMCPmc4136000.
    1. de Wilde AH, Jochmans D, Posthuma CC, Zevenhoven-Dobbe JC, van Nieuwkoop S, Bestebroer TM, et al. Screening of an FDA-approved compound library identifies four small-molecule inhibitors of Middle East respiratory syndrome coronavirus replication in cell culture. Antimicrobial agents and chemotherapy. 2014;58(8):4875–84. doi: ; PubMed Central PMCID: PMC4136071.
    1. Cong Y, McArthur MA, Cohen M, Jahrling PB, Janosko KB, Josleyn N, et al. Characterization of yellow fever virus infection of human and non-human primate antigen presenting cells and their Interaction with CD4+ T Cells. PLoS Negl Trop Dis. 2016;10(5):e0004709 doi: ; PubMed Central PMCID: PMC4871483.
    1. Sandow D, Hofmann F, Gropler K, Nuhn P. [Statistical analysis of the in-vitro action of combination antiviral agents]. Z Gesamte Hyg. 1990;36(9):471–4. .
    1. Wulff NH, Tzatzaris M, Young PJ. Monte Carlo simulation of the Spearman-Kaerber TCID50. J Clin Bioinforma. 2012;2(1):5 doi: ; PubMed Central PMCID: PMC3331834.
    1. Graham RL, Donaldson EF, Baric RS. A decade after SARS: strategies for controlling emerging coronaviruses. Nature reviews Microbiology. 2013;11(12):836–48. Epub 2013/11/13. doi: .
    1. Chu H, Zhou J, Wong BH, Li C, Cheng ZS, Lin X, et al. Productive replication of Middle East respiratory syndrome coronavirus in monocyte-derived dendritic cells modulates innate immune response. Virology. 2014;454–455:197–205. doi: .
    1. Fugier-Vivier I, Servet-Delprat C, Rivailler P, Rissoan MC, Liu YJ, Rabourdin-Combe C. Measles virus suppresses cell-mediated immunity by interfering with the survival and functions of dendritic and T cells. J Exp Med. 1997;186(6):813–23. ; PubMed Central PMCID: PMC2199042.
    1. Granelli-Piperno A, Finkel V, Delgado E, Steinman RM. Virus replication begins in dendritic cells during the transmission of HIV-1 from mature dendritic cells to T cells. Curr Biol. 1999;9(1):21–9. .
    1. Rescigno M, Winzler C, Delia D, Mutini C, Lutz M, Ricciardi-Castagnoli P. Dendritic cell maturation is required for initiation of the immune response. Journal of leukocyte biology. 1997;61(4):415–21. .
    1. Banchereau J, Briere F, Caux C, Davoust J, Lebecque S, Liu YJ, et al. Immunobiology of dendritic cells. Annu Rev Immunol. 2000;18:767–811. doi: .
    1. Hart DN. Dendritic cells: unique leukocyte populations which control the primary immune response. Blood. 1997;90(9):3245–87. .
    1. Sallusto F, Lanzavecchia A. Efficient presentation of soluble antigen by cultured human dendritic cells is maintained by granulocyte/macrophage colony-stimulating factor plus interleukin 4 and downregulated by tumor necrosis factor alpha. J Exp Med. 1994;179(4):1109–18. ; PubMed Central PMCID: PMC2191432.
    1. Spiegel M, Schneider K, Weber F, Weidmann M, Hufert FT. Interaction of severe acute respiratory syndrome-associated coronavirus with dendritic cells. The Journal of general virology. 2006;87(Pt 7):1953–60. Epub 2006/06/09. doi: .
    1. Kruse M, Rosorius O, Kratzer F, Stelz G, Kuhnt C, Schuler G, et al. Mature dendritic cells infected with herpes simplex virus type 1 exhibit inhibited T-cell stimulatory capacity. Journal of virology. 2000;74(15):7127–36. ; PubMed Central PMCID: PMC112231.
    1. Alloatti A, Kotsias F, Magalhaes JG, Amigorena S. Dendritic cell maturation and cross-presentation: timing matters! Immunol Rev. 2016;272(1):97–108. doi: .
    1. Drosten C, Seilmaier M, Corman VM, Hartmann W, Scheible G, Sack S, et al. Clinical features and virological analysis of a case of Middle East respiratory syndrome coronavirus infection. Lancet Infect Dis. 2013;13(9):745–51. Epub 2013/06/21. doi: .
    1. Huang KJ, Su IJ, Theron M, Wu YC, Lai SK, Liu CC, et al. An interferon-gamma-related cytokine storm in SARS patients. Journal of medical virology. 2005;75(2):185–94. Epub 2004/12/17. doi: .
    1. Nicholls JM, Poon LL, Lee KC, Ng WF, Lai ST, Leung CY, et al. Lung pathology of fatal severe acute respiratory syndrome. Lancet. 2003;361(9371):1773–8. .
    1. Zhou J, Chu H, Li C, Wong BH, Cheng ZS, Poon VK, et al. Active replication of Middle East respiratory syndrome coronavirus and aberrant induction of inflammatory cytokines and chemokines in human macrophages: implications for pathogenesis. The Journal of infectious diseases. 2014;209(9):1331–42. Epub 2013/09/26. doi: .
    1. Jiang Y, Xu J, Zhou C, Wu Z, Zhong S, Liu J, et al. Characterization of cytokine/chemokine profiles of severe acute respiratory syndrome. American journal of respiratory and critical care medicine. 2005;171(8):850–7. Epub 2005/01/20. doi: .
    1. Tynell J, Westenius V, Ronkko E, Munster VJ, Melen K, Osterlund P, et al. Middle East respiratory syndrome coronavirus shows poor replication but significant induction of antiviral responses in human monocyte-derived macrophages and dendritic cells. The Journal of general virology. 2016;97(2):344–55. doi: ; PubMed Central PMCID: PMC4804640.
    1. Scheuplein VA, Seifried J, Malczyk AH, Miller L, Hocker L, Vergara-Alert J, et al. High secretion of interferons by human plasmacytoid dendritic cells upon recognition of Middle East respiratory syndrome coronavirus. Journal of virology. 2015;89(7):3859–69. doi: ; PubMed Central PMCID: PMC4403407.
    1. Bergamaschi A, Pancino G. Host hindrance to HIV-1 replication in monocytes and macrophages. Retrovirology. 2010;7:31 doi: ; PubMed Central PMCID: PMC2868797.
    1. Josset L, Menachery VD, Gralinski LE, Agnihothram S, Sova P, Carter VS, et al. Cell host response to infection with novel human coronavirus EMC predicts potential antivirals and important differences with SARS coronavirus. MBio. 2013;4(3):e00165–13. doi: ; PubMed Central PMCID: PMC3663187.
    1. Johansen LM, Brannan JM, Delos SE, Shoemaker CJ, Stossel A, Lear C, et al. FDA-approved selective estrogen receptor modulators inhibit Ebola virus infection. Sci Transl Med. 2013;5(190):190ra79 doi: ; PubMed Central PMCID: PMC3955358.
    1. Thome R, Moraes AS, Bombeiro AL, Farias Ados S, Francelin C, da Costa TA, et al. Chloroquine treatment enhances regulatory T cells and reduces the severity of experimental autoimmune encephalomyelitis. PLoS One. 2013;8(6):e65913 doi: ; PubMed Central PMCID: PMC3683039.
    1. Savarino A, Boelaert JR, Cassone A, Majori G, Cauda R. Effects of chloroquine on viral infections: an old drug against today's diseases? Lancet Infect Dis. 2003;3(11):722–7. .
    1. Akpovwa H. Chloroquine could be used for the treatment of filoviral infections and other viral infections that emerge or emerged from viruses requiring an acidic pH for infectivity. Cell Biochem Funct. 2016;34(4):191–6. doi: ; PubMed Central PMCID: PMC5071688.
    1. Long J, Wright E, Molesti E, Temperton N, Barclay W. Antiviral therapies against Ebola and other emerging viral diseases using existing medicines that block virus entry. F1000Res. 2015;4:30 doi: ; PubMed Central PMCID: PMC4431382.
    1. Madrid PB, Chopra S, Manger ID, Gilfillan L, Keepers TR, Shurtleff AC, et al. A systematic screen of FDA-approved drugs for inhibitors of biological threat agents. PLoS One. 2013;8(4):e60579 doi: ; PubMed Central PMCID: PMC3618516.
    1. Porotto M, Orefice G, Yokoyama CC, Mungall BA, Realubit R, Sganga ML, et al. Simulating henipavirus multicycle replication in a screening assay leads to identification of a promising candidate for therapy. Journal of virology. 2009;83(10):5148–55. doi: ; PubMed Central PMCID: PMC2682105.
    1. Miyamoto S, Miyake N, Jarskog LF, Fleischhacker WW, Lieberman JA. Pharmacological treatment of schizophrenia: a critical review of the pharmacology and clinical effects of current and future therapeutic agents. Mol Psychiatry. 2012;17(12):1206–27. doi: .
    1. Chu JJ, Ng ML. Infectious entry of West Nile virus occurs through a clathrin-mediated endocytic pathway. Journal of virology. 2004;78(19):10543–55. doi: ; PubMed Central PMCID: PMC516396.

Source: PubMed

3
订阅