Chemokine up-regulation in SARS-coronavirus-infected, monocyte-derived human dendritic cells

Helen K W Law, Chung Yan Cheung, Hoi Yee Ng, Sin Fun Sia, Yuk On Chan, Winsie Luk, John M Nicholls, J S Malik Peiris, Yu Lung Lau, Helen K W Law, Chung Yan Cheung, Hoi Yee Ng, Sin Fun Sia, Yuk On Chan, Winsie Luk, John M Nicholls, J S Malik Peiris, Yu Lung Lau

Abstract

Lymphopenia and increasing viral load in the first 10 days of severe acute respiratory syndrome (SARS) suggested immune evasion by SARS-coronavirus (CoV). In this study, we focused on dendritic cells (DCs) which play important roles in linking the innate and adaptive immunity. SARS-CoV was shown to infect both immature and mature human monocyte-derived DCs by electron microscopy and immunofluorescence. The detection of negative strands of SARS-CoV RNA in DCs suggested viral replication. However, no increase in viral RNA was observed. Using cytopathic assays, no increase in virus titer was detected in infected DCs and cell-culture supernatant, confirming that virus replication was incomplete. No induction of apoptosis or maturation was detected in SARS-CoV-infected DCs. The SARS-CoV-infected DCs showed low expression of antiviral cytokines (interferon alpha [IFN-alpha], IFN-beta, IFN-gamma, and interleukin 12p40 [IL-12p40]), moderate up-regulation of proinflammatory cytokines (tumor necrosis factor alpha [TNF-alpha] and IL-6) but significant up-regulation of inflammatory chemokines (macrophage inflammatory protein 1alpha [MIP-1alpha], regulated on activation normal T cell expressed and secreted [RANTES]), interferon-inducible protein of 10 kDa [IP-10], and monocyte chemoattractant protein 1 [MCP-1]). The lack of antiviral cytokine response against a background of intense chemokine up-regulation could represent a mechanism of immune evasion by SARS-CoV.

Figures

Figure 1
Figure 1
Electron microscopy of SARS-CoV–infected human DCs. Negative-contrast thin-section transmission electron micrograph of SARS-CoV–infected human DCs showed virus (black arrowheads) binding (A) and uptake (B) at 0 hours after infection. At 6 hours, 12 hours, and 24 hours after infection, virus particles were detected in endosomes (C) and cytoplasm (D) but not in the Golgi apparatus (E). No virus budding was observed (F) in all cells examined. Images are representative of SARS-CoV–infected human DCs from 3 independent adult or CB donors.
Figure 2
Figure 2
Immunofluorescence assay for SARS-CoV detection in human DCs. Mock-infected human DCs were included as a control (A). Positive immunofluorescence staining was detected in human immature and mature DCs at 12 hours (B) and 24 hours (C) after infection with SARS-CoV (MOI = 1). Confocal microscopy showed positive staining in the cytoplasm of DCs (D). Images are representative of immature and mature DCs from 11 independent adult or CB donors.
Figure 3
Figure 3
Viral gene expression in SARS-CoV–infected human adult immature DCs by quantitative RT-PCR. Both negative (A) and positive strands (B) of SARS-CoV Replicase 1b mRNA were detected in SASR-CoV–infected adult immature DCs at 3 hours, 9 hours, 24 hours, day 3, and day 6 after infection (MOI = 1). Similar pattern of decreased viral gene expression was detected for the negative and positive strands in all 3 cases.
Figure 4
Figure 4
Active caspase-3 assay for apoptosis in human adult immature DCs. Comparing with mock-infected adult immature DCs, no significant induction of active caspase-3–positive cells was observed at 6 hours, 12 hours, and 24 hours after infection (n = 4; P > .05). Data are shown as mean ± SEM of DCs from 4 independent donors. The percentage of active caspase-3–positive Jurkat cells in the positive control at 3 hours and 24 hours after addition of anti-Fas antibodies were 15% and 35%, respectively.
Figure 5
Figure 5
Flow cytometry analysis of cell-surface molecule expression on human adult DCs. Mock- (A) or SARS-CoV–infected (B) adult immature DCs (MOI = 1) were harvested at 48 hours after infection and stained for flow cytometry analysis. Surface staining is shown by filled histogram, and isotype control is marked by the dotted line. SARS-CoV alone did not up-regulate the expression of CD83, CD86, MHC class I, and MHC class II. However, SARS-CoV–infected cells can be stimulated by LPS (10 μg/mL; thick line) to up-regulate the expression of these molecules to similar levels as in the mock-infected controls. Data shown are representative of adult immature DCs from 5 independent donors. FSC indicates forward scatter; SSC, side scatter.
Figure 6
Figure 6
Antiviral cytokine gene expression profile of SARS-CoV–infected human immature DCs by quantitative RT-PCR. Antiviral cytokine mRNA concentrations in adult (A) and CB (B) immature cells were assayed at 3 hours and 9 hours after infection with SARS-CoV (MOI = 1). Mock-infected cells were included as negative control. The concentrations were normalized to those of β-actin mRNA in the corresponding sample. There were low expressions of IFN-α, IFN-β, IFN-γ, and IL-12p40 genes in SARS-CoV–infected DCs. Data are shown as mean ± SEM (adult n = 7; CB n = 5).
Figure 7
Figure 7
Proinflammatory cytokine gene expression profile of SARS-CoV–infected human immature DCs by quantitative RT-PCR. Proinflammatory cytokine mRNA concentrations in adult (A) and CB (B) immature cells were assayed at 3 hours and 9 hours after infection with SARS-CoV (MOI = 1). Mock-infected cells were included as negative control. The concentrations were normalized to those of β-actin mRNA in the corresponding sample. There were moderate up-regulation of TNF-α and IL-6 expression in SARS-CoV–infected DCs. Data are shown as mean ± SEM (adult n = 7; CB n = 5; *P < .05).
Figure 8
Figure 8
Chemokine gene expression profile of SARS-CoV–infected human immature DCs by quantitative RT-PCR. Chemokine mRNA concentrations in adult (A) and CB (B) immature cells were assayed at 3 hours and 9 hours after infection with SARS-CoV (MOI = 1). Mock-infected cells were included as negative control. The concentrations were normalized to those of β-actin mRNA in the corresponding sample. There was significant up-regulation of MIP-1α, RANTES, IP-10, and MCP-1 in SARS-CoV–infected DCs. Data are shown as mean ± SEM (adult n = 7; CB n = 5; *P < .05).

References

    1. Lai MMC, Holmes KV. Fields' virology. Lippincott Williams & Wilkins; Philadelphia, PA: 2001. Coronaviridae: the viruses and their replication. In: Knipe DM, Howley PM, eds. pp. 1163–1185.
    1. Choi KW, Chau TN, Tsang O, et al. Outcomes and prognostic factors in 267 patients with severe acute respiratory syndrome in Hong Kong. Ann Intern Med. 2003;139:715–723.
    1. Peiris JS, Lai ST, Poon LL, et al. Coronavirus as a possible cause of severe acute respiratory syndrome. Lancet. 2003;361:1319–1325.
    1. Peiris JS, Chu CM, Cheng VC, et al. Clinical progression and viral load in a community outbreak of coronavirus-associated SARS pneumonia: a prospective study. Lancet. 2003;361:1767–1772.
    1. Drosten C, Gunther S, Preiser W, et al. Identification of a novel coronavirus in patients with severe acute respiratory syndrome. N Engl J Med. 2003;348:1967–1976.
    1. Wong RS, Wu A, To KF, et al. Haematological manifestations in patients with severe acute respiratory syndrome: retrospective analysis. BMJ. 2003;326:1358–1362.
    1. Nicholls JM, Poon LL, Lee KC, et al. Lung pathology of fatal severe acute respiratory syndrome. Lancet. 2003;361:1773–1778.
    1. Ding Y, Wang H, Shen H, et al. The clinical pathology of severe acute respiratory syndrome (SARS): a report from China. J Pathol. 2003;200:282–289.
    1. Banchereau J, Briere F, Caux C, et al. Immunobiology of dendritic cells. Annu Rev Immunol. 2000;18:767–811.
    1. Palucka K, Banchereau J. Dendritic cells: a link between innate and adaptive immunity. J Clin Immunol. 1999;19:12–25.
    1. Lipscomb MF, Masten BJ. Dendritic cells: immune regulators in health and disease. Physiol Rev. 2002;82:97–103.
    1. Peebles RS, Jr, Graham BS. Viruses, dendritic cells and the lung. Respir Res. 2001;2:245–249.
    1. Legge KL, Braciale TJ. Accelerated migration of respiratory dendritic cells to the regional lymph nodes is limited to the early phase of pulmonary infection. Immunity. 2003;18:265–277.
    1. Engering A, Geijtenbeek TB, van Kooyk Y. Immune escape through C-type lectins on dendritic cells. Trends Immunol. 2002;23:480–485.
    1. Geijtenbeek TB, Engering A, Van Kooyk Y. DC-SIGN, a C-type lectin on dendritic cells that unveils many aspects of dendritic cell biology. J Leukoc Biol. 2002;71:921–931.
    1. Barton GM, Medzhitov R. Toll-like receptors and their ligands. Curr Top Microbiol Immunol. 2002;270:81–92.
    1. Barton GM, Medzhitov R. Toll-like receptor signaling pathways. Science. 2003;300:1524–1525.
    1. Steinman RM, Pope M. Exploiting dendritic cells to improve vaccine efficacy. J Clin Invest. 2002;109:1519–1526.
    1. Dieu-Nosjean MC, Vicari A, Lebecque S, Caux C. Regulation of dendritic cell trafficking: a process that involves the participation of selective chemokines. J Leukoc Biol. 1999;66:252–262.
    1. Sozzani S, Allavena P, Vecchi A, Mantovani A. Chemokines and dendritic cell traffic. J Clin Immunol. 2000;20:151–160.
    1. Glass WG, Rosenberg HF, Murphy PM. Chemokine regulation of inflammation during acute viral infection. Curr Opin Allergy Clin Immunol. 2003;3:467–473.
    1. Liu E, Tu W, Law HK, Lau YL. Decreased yield, phenotypic expression and function of immature monocyte-derived dendritic cells in cord blood. Br J Haematol. 2001;113:240–246.
    1. Liu E, Tu W, Law HK, Lau YL. Changes of CD14 and CD1a expression in response to IL-4 and granulocyte-macrophage colony-stimulating factor are different in cord blood and adult blood monocytes. Pediatr Res. 2001;50:184–189.
    1. Liu E, Law HK, Lau YL. BCG promotes cord blood monocyte-derived dendritic cell maturation with nuclear Rel-B up-regulation and cytosolic I kappa B alpha and beta degradation. Pediatr Res. 2003;54:105–112.
    1. Liu E, Law HK, Lau YL. Insulin-like growth factor I promotes maturation and inhibits apoptosis of immature cord blood monocyte-derived dendritic cells through MEK and PI 3-kinase pathways. Pediatr Res. 2003;54:919–925.
    1. Liu E, Law HK, Lau YL. Tolerance associated with cord blood transplantation may depend on the state of host dendritic cells. Br J Haematol. 2004;126:517–526.
    1. Cheung CY, Poon LL, Lau AS, et al. Induction of proinflammatory cytokines in human macrophages by influenza A (H5N1) viruses: a mechanism for the unusual severity of human disease? Lancet. 2002;360:1831–1837.
    1. Li L, Wo J, Shao J, et al. SARS-coronavirus replicates in mononuclear cells of peripheral blood (PBMCs) from SARS patients. J Clin Virol. 2003;28:239–244.
    1. Lau YL. SARS: future research and vaccine. Paediatr Respir Rev. 2004;5:300–303.
    1. Sieczkarski SB, Whittaker GR. Dissecting virus entry via endocytosis. J Gen Virol. 2002;83:1535–1545.
    1. Yeager CL, Ashmun RA, Williams RK, et al. Human aminopeptidase N is a receptor for human coronavirus 229E. Nature. 1992;357:420–422.
    1. van der Velden VH, Leenen PJ, Drexhage HA. CD13/aminopeptidase N involvement in dendritic cell maturation. Leukemia. 2001;15:190–191.
    1. Rosenzwajg M, Tailleux L, Gluckman JC. CD13/N-aminopeptidase is involved in the development of dendritic cells and macrophages from cord blood CD34(+) cells. Blood. 2000;95:453–460.
    1. Li W, Moore MJ, Vasilieva N, et al. Angiotensin-converting enzyme 2 is a functional receptor for the SARS coronavirus. Nature. 2003;426:450–454.
    1. Hamming I, Timens W, Bulthuis ML, Lely AT, Navis GJ, van Goor H. Tissue distribution of ACE2 protein, the functional receptor for SARS coronavirus. A first step in understanding SARS pathogenesis. J Pathol. 2004;203:631–637.
    1. To KF, Lo AW. Exploring the pathogenesis of severe acute respiratory syndrome (SARS): the tissue distribution of the coronavirus (SARS-CoV) and its putative receptor, angiotensin-converting enzyme 2 (ACE2). J Pathol. 2004;203:740–743.
    1. Rota PA, Oberste MS, Monroe SS, et al. Characterization of a novel coronavirus associated with severe acute respiratory syndrome. Science. 2003;300:1394–1399.
    1. Krokhin O, Li Y, Andonov A, et al. Mass spectrometric characterization of proteins from the SARS virus: a preliminary report. Mol Cell Proteomics. 2003;2:346–356.
    1. Yang ZY, Huang Y, Ganesh L, et al. pH-dependent entry of severe acute respiratory syndrome coronavirus is mediated by the spike glycoprotein and enhanced by dendritic cell transfer through DC-SIGN. J Virol. 2004;78:5642–5650.
    1. Ksiazek TG, Erdman D, Goldsmith CS, et al. A novel coronavirus associated with severe acute respiratory syndrome. N Engl J Med. 2003;348:1953–1966.
    1. Collins AR. In vitro detection of apoptosis in monocytes/macrophages infected with human coronavirus. Clin Diagn Lab Immunol. 2002;9:1392–1395.
    1. Schneider-Schaulies S, Klagge IM, ter Meulen V. Dendritic cells and measles virus infection. Curr Top Microbiol Immunol. 2003;276:77–101.
    1. Marrack P, Kappler J, Mitchell T. Type I interferons keep activated T cells alive. J Exp Med. 1999;189:521–530.
    1. Cella M, Salio M, Sakakibara Y, Langen H, Julkunen I, Lanzavecchia A. Maturation, activation, and protection of dendritic cells induced by double-stranded RNA. J Exp Med. 1999;189:821–829.
    1. Seet BT, Johnston JB, Brunetti CR, et al. Poxviruses and immune evasion. Annu Rev Immunol. 2003;21:377–423.
    1. Kadowaki N, Ho S, Antonenko S, et al. Subsets of human dendritic cell precursors express different toll-like receptors and respond to different microbial antigens. J Exp Med. 2001;194:863–869.
    1. Levy DE, Marie L, Prakash A. Ringing the interferon alarm: differential regulation of gene expression at the interface between innate and adaptive immunity. Curr Opin Immunol. 2003;15:52–58.
    1. Le Bon S, Schiavoni G, D'Agostino G, Gresser I, Belardelli F, Tough DF. Type I interferons potently enhance humoral immunity and can promote isotype switching by stimulating dendritic cells in vivo. Immunity. 2001;14:461–470.
    1. Watford WT, Moriguchi M, Morinobu A. O'Shea JJ. The biology of IL-12: coordinating innate and adaptive immune responses. Cytokine Growth Factor Rev. 2003;14:361–368.
    1. Tassaneetrithep B, Burgess TH, Granelli-Piperno A, et al. DC-SIGN (CD209) mediates dengue virus infection of human dendritic cells. J Exp Med. 2003;197:823–829.
    1. Locksley RM, Killeen N, Lenardo MJ. The TNF receptor superfamilies: integrating mammalian biology. Cell. 2001;104:487–501.
    1. Ho JC, Ooi GC, Mok TY, et al. High-dose pulse versus nonpulse corticosteroid regimens in severe acute respiratory syndrome. Am J Respir Crit Care Med. 2003;168:1449–1456.
    1. Rot A, von Andrian UH. Chemokines in innate and adaptive host defense: basic chemokinese grammar for immune cells. Annu Rev Immunol. 2004;22:891–928.
    1. Mahalingam S, Friedland JS, Heise MT, Rulli NE, Meanger J, Lidbury BA. Chemokines and viruses: friends or foes? Trends Microbiol. 2003;11:383–391.
    1. Wong CK, Lam CW, Wu AK, et al. Plasma inflammatory cytokines and chemokines in severe acute respiratory syndrome. Clin Exp Immunol. 2004;136:95–103.
    1. Glass WG, Subbarao K, Murphy B, Murphy PM. Mechanisms of host defense following severe acute respiratory syndrome-coronavirus (SARS-CoV) pulmonary infection of mice. J Immunol. 2004;173:4030–4039.
    1. Kaufmann A, Salentin R, Meyer RG, et al. Defense against influenza A virus infection: essential role of the chemokine system. Immunobiology. 2001;204:603–613.
    1. Sallusto F, Lanzavecchia A. Understanding dendritic cell and T-lymphocyte traffic through the analysis of chemokine receptor expression. Immunol Rev. 2000;177:134–140.
    1. Larsson M, Beignon AS, Bhardwaj N. DC-virus interplay: a double edged sword. Semin Immunol. 2004;16:147–161.

Source: PubMed

3
Abonnieren