Low levels of antibody-dependent enhancement in vitro using viruses and plasma from dengue patients

Panjaporn Chaichana, Tamaki Okabayashi, Orapim Puiprom, Mikiko Sasayama, Tadahiro Sasaki, Akifumi Yamashita, Pongrama Ramasoota, Takeshi Kurosu, Kazuyoshi Ikuta, Panjaporn Chaichana, Tamaki Okabayashi, Orapim Puiprom, Mikiko Sasayama, Tadahiro Sasaki, Akifumi Yamashita, Pongrama Ramasoota, Takeshi Kurosu, Kazuyoshi Ikuta

Abstract

Background: The majority of dengue patients infected with any serotype of dengue virus (DENV) are asymptomatic, but the remainder may develop a wide spectrum of clinical symptoms, ranging from mild dengue fever (DF) to severe dengue hemorrhagic fever (DHF). Severe cases occur more often in patients who experience a secondary infection with a different virus serotype. A phenomenon called antibody-dependent enhancement (ADE) has been proposed to explain the onset of these severe cases, but the exact mechanism of ADE remains unclear.

Methodology/principal finding: Virus neutralization and ADE assays were performed using ultracentrifugation supernatants of acute-phase sera from patients with secondary infections or human monoclonal antibodies (HuMAbs) as anti-DENV antibodies. Virus sources included infectious serum-derived viruses from the ultracentrifugation precipitates, laboratory-culture adapted DENV, or recombinant DENVs derived from patient sera. In contrast to the high levels of ADE observed with laboratory virus strains, low ADE was observed with autologous patient-derived viruses, when patient sera were used to provide the antibody component in the ADE assays. Similar results were obtained using samples from DF and DHF patients. Recombinant-viruses derived from DHF patients showed only minor differences in neutralization and ADE activity in the presence of HuMAbs or plasma derived from the same DHF patient.

Conclusion/significance: Serum or plasma taken from patients during the acute phase of a secondary infection showed high levels of ADE, but no neutralization activity, when assayed in the presence of laboratory-adapted virus strains. By contrast, serum or plasma from the same patient showed high levels of neutralization activity but failed to induce significant ADE when the assays were performed with autologous virus. These results demonstrate the significance of the virus source when measuring ADE. They also suggest that repeated passage of DENV in cell culture has endowed it with the capacity to induce high levels of ADE.

Conflict of interest statement

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

Figures

Figure 1. Construction of recombinant DENVs based…
Figure 1. Construction of recombinant DENVs based on virus samples from patient plasma.
RNA was extracted from the plasma of patient D30 and the prM-E encoding region of DENV was amplified by RT-PCR and cloned into the pCR4Blunt-TOPO vector for DNA sequence analysis. Representative prM-E region variants were selected and used to construct full-length DENV cDNA clones based on plasmid pmMW/R05-624. The resultant full-length cDNAs containing D30-derived variants were used as templates for RNA synthesis, and in vitro-transcribed viral RNAs were transfected into C6/36 cells. Supernatants from transfected cells were passaged once in C6/36 to obtain adequate quantities of viruses. EDI, envelope domain I. EDII, envelope domain II. EDIII, envelope domain III. TM, transmembrane. The recombinant DENV clone in brackets was excluded from further study due to an inadequate titer.
Figure 2. Low ADE when using serum…
Figure 2. Low ADE when using serum and viruses from DENV-patients.
Serum samples (DENV-1, DENV-2, and DENV-3) from HTD dengue patients with DF and DHF symptoms were ultracentrifuged to precipitate DENV virions which were used in assays without any subsequent passage in cells. The supernatant fractions were heat-inactivated at 56°C for 30 minutes and then serially diluted 10-fold. The dilutions were mixed for 30 minutes at 37°C with the precipitated virions from autologous plasma at an MOI of 0.02. The virus-antibody complexes were added to K562 cells and incubated for 2 hours at 37°C before the addition of maintenance medium supplemented with 2% FBS. The cells were then incubated for a further 3 days. Supernatants were harvested for virus titration by focus-forming immunoassay in Vero cells, and the results are expressed as FFU/ml. The mean ± SD of triplicate experiment is shown. ‘No Ab’ means virus infection in the absence of plasma. The ‘No Ab’ value was used as a baseline for calculating virus infection enhancement.
Figure 3. High levels of ADE when…
Figure 3. High levels of ADE when using patient plasma and laboratory-culture adapted DENV.
Ultracentrifugation supernatants of patient sera were heat-inactivated at 56°C for 30 min, diluted 10-fold, and pre-mixed at an MOI of 0.02 with laboratory culture-adapted DENV-1, -2, and -3 for 30 minutes at 37°C. Virus-antibody complexes were added to K562 cells and incubated for 2 hours at 37°C. Maintenance medium supplemented with 2% FBS was then added before a further incubation for 3 days. Supernatants were harvested for virus titration in focus-forming immunoassays in Vero cells. Results are expressed as the mean ± SD of triplicate experiments. ‘No Ab’ means virus infection in the absence of plasma. The ‘No Ab’ value was used as a base line for calculating virus infection enhancement.
Figure 4. Replication kinetics of recombinant DENVs.
Figure 4. Replication kinetics of recombinant DENVs.
DENV-2 16681 or individual recombinant DENVs were used to infect C6/36 (A) and Vero cells (B) at an MOI of 0.001, or K562 cells (C) at an MOI of 0.1. After 2 hours of incubation, the supernatants were removed and cells were washed twice with plain medium before the addition of maintenance medium supplemented with 2% FBS. For infected C6/36 and Vero cells, the supernatants were harvested daily. For infected K562 cells, both the culture medium and infected cells were harvested and centrifuged. Virus titers in the supernatants were determined in focus-forming assays in Vero cells. Results are expressed as mean ± SD of triplicate experiment (*p<0.05 and **p<0.01, unpaired two-tailed Student’s t-test, n = 3 per point). Statistically significant differences between data points are indicated by # (# p<0.05, ## p<0.01).
Figure 5. ADE levels of recombinant DENVs…
Figure 5. ADE levels of recombinant DENVs or DENV-2 strain 16681 exposed to D30-plasma or HuMAbs.
A heat-inactivated D30-plasma sample (A) or purified human MAbs derived from patient D30 (B) were serially diluted 10-fold in RPMI-1640 medium and incubated with either DENV-2 16681 or individual recombinant DENVs for 30 min at an MOI of 0.1. Virus-antibody complexes were added to K562 cells and incubated for a further 2 h; maintenance medium was then added (without washing the cells) to yield a FBS final concentration of 2%. Cells and supernatants were collected on Day 3 post-infection. Virus titers in the supernatants were determined in focus-forming immunoassays in Vero cells. Results are expressed as the mean ± SD from two independent experiments performed in triplicate (*p<0.05 and **p<0.01, unpaired two-tailed Student t-test, n = 3 per point). Statistically significant differences between data points are indicated by # (# p<0.05, ## p<0.01).

References

    1. Kuhn RJ, Zhang W, Rossmann MG, Platnev SV, Corver J, et al. (2002) Structure of dengue virus: implications for flavivirus organization, maturation, and fusion. Cell 108: 717–725.
    1. van der Schaar HM, Wilschut JC, Smit JM (2009) Role of antibodies in controlling dengue virus infection. Immunobiology 214: 613–629.
    1. Midgley CM, Bajwa-Joseph M, Vasanawathana S, Limpitikul W, Wills B, et al. (2011) An in-depth analysis of original antigenic sin in dengue virus infection. J Virol 85: 410–421.
    1. Sangkawibha N, Rojanasuphot S, Ahandrik S, Viriyapongse S, Jatanasen S, et al. (1984) Risk factors in dengue shock syndrome: a prospective epidemiologic study in Rayong, Thailand. I. The 1980 outbreak. Am J Epidemiol 120: 653–669.
    1. Guzmán MG, Kouri GP, Bravo J, Soler M, Vazquez S, et al. (1990) Dengue hemorrhagic fever in Cuba, 1981: a retrospective seroepidemiologic study. Am J Trop Med Hyg 42: 179–184.
    1. Rothman AL (2010) Cellular immunology of sequential dengue virus infection and its role in disease pathogenesis. Curr Top Microbiol Immunol 338: 83–98.
    1. Halstead SB, O’Rourke EJ (1977) Dengue viruses and mononuclear phagocytes. I. Infection enhancement by non-neutralizing antibody. J Exp Med 146: 201–217.
    1. Sabin AB (1952) Research on dengue during World War II. Am J Trop Med Hyg 1: 30–50.
    1. García G, Sierra B, Pérez AB, Aguirre E, Rosado I, et al. (2010) Asymptomatic dengue infection in a Cuban population confirms the protective role of the RR variant of the FcγRIIa polymorphism. Am J Trop Med Hyg 82: 1153–1156.
    1. Libraty DH, Acosta LP, Tallo V, Segubre-Mercado E, Bautista A, et al. (2009) A prospective nested case-control study of dengue in infects: Rethinking and refining the antibody-dependent enhancement dengue hemorrhagic fever model. PloS Med 6: e1000171.
    1. Balsitis SJ, Coloma J, Castro G, Alava A, Flores D, et al. (2009) Tropism of dengue virus in mice and human defined by viral nonstructural protein 3-specific immunostaining. Am J Trop Med Hyg 80: 416–424.
    1. Jessie K, Fong MY, Devi S, Lam SK, Wong KT (2004) Localization of dengue virus in naturally infected human tissues, by immunohistochemistry and in situ hybridization. J Infect Dis 189: 1411–1418.
    1. Baclig MO, Gervacio LT, Suarez LA, Buerano CC, Matias RR, et al. (2010) Flow cytometric analysis of dengue virus-infected cells in peripheral blood. Southeast Asian J Trop Med Public Health 41: 1352–1358.
    1. Durbin AP, Vargas MJ, Wanionek K, Hammond SN, Gordon A, et al. (2008) Phenotyping of peripheral blood mononuclear cells during acute dengue illness demonstrates infection and increased activation of monocytes in severe cases compared to classic dengue fever. Virology 376: 429–435.
    1. King AD, Nisalak A, Kalayanrooj S, Myint KS, Pattanapanyasat K, et al. (1999) B cells are the principal mononuclear cells infected by dengue virus. Southeast Asian J Trop Med Public Health 30: 718–728.
    1. Srikiatkhachorn A, Wichit S, Gibbons RV, Green S, Libraty DH, et al. (2012) Dengue viral RNA levels in peripheral blood mononuclear cells are associated with disease severity and pre-existing dengue immune status. PloS One 7: e51335.
    1. Huang KJ, Yang YC, Lin YS, Huang JH, Liu HS, et al. (2006) The dual-specific binding of dengue virus and target cells for the antibody-dependent enhancement of dengue virus infection. J Immunol 176: 2825–2832.
    1. Dejnirattisai W, Jumnainsong A, Onsirisakul N, Fitton P, Vasanawathana S, et al. (2010) Cross-reacting antibodies enhance dengue virus infection in humans. Science 328: 745–748.
    1. Beltramello M, Williams KL, Simmons CP, Macagno A, Simonelli L, et al. (2010) The human immune response to dengue virus is dominated by highly cross-reactive antibodies endowed with neutralizing and enhancing activity. Cell Host Microbe 8: 271–183.
    1. Rodenhuis-Zybert I, Moesker B, de Silva Voorham JM, der Ende-Metselaar H, Diamond MS, et al. (2011) A fusion-loop antibody enhances the infectious properties of immature flavivirus particles. J Virol 85: 11800–11808.
    1. Hughes HR, Crill WD, Chang GJ (2012) Manipulation of immunodominant dengue virus E protein epitopes reduces potential antibody-dependent enhancement. Virol J 9: 115.
    1. da Silva Voorham JM, Rodenhuis-Zybert IA, Ayala Nunez NV, Colpitts TM, van der Ende-Metselaar H, et al. (2012) Antibodies against the envelope glycoprotein promote infectivity of immature dengue virus serotype 2. PloS One 7: e29957.
    1. Sasaki T, Setthapramote C, Kurosu T, Nishimura M, Asai A, et al... (2013) Dengue virus neutralization and antibody-dependent enhancement activities of human monoclonal antibodies derived from dengue patients at acute phase of secondary infection. Antiviral Res 98: 423–431, 2013.
    1. Guy B, Chanthavanich P, Gimenez S, Sirivichayakul C, Sabchareon A, et al. (2004) Evaluation by flow cytometry of antibody-dependent enhancement (ADE) of dengue infection by sera from Thai children immunized a live-attenuated tetravalent dengue vaccine. Vaccine 22: 3563–74.
    1. Guy B, Barrere B, Malinowski C, Saville M, Teyssou R, et al. (2011) From research to phase III: preclinical, industrial and clinical development of the Sanofi Pasteur tetravalent dengue vaccine. Vaccine 29: 7229–7241.
    1. Konishi E, Tabuchi Y, Yamanaka A (2010) A simple assay system for infection-enhancing and –neutralizing antibodies to dengue type 2 virus using layers of semi-adherent K562 cells. J Virol Methods 163: 360–367.
    1. Halstead SB, Simasthien P (1970) Observations related to the pathogenesis of dengue hemorrhagic fever. II. Antigenic and biologic properties of dengue viruses and their association with disease response in the host. Yale J Biol Med 42(5): 276–92.
    1. Kubota-Koketsu R, Mizuta H, Oshita M, Ideno S, Yunoki M, et al. (2009) Broad neutralizing human monoclonal antibodies against influenza virus from vaccinated healthy donors. Biochem Biophys Res Commun 387: 180–185.
    1. Falconar AK (1999) Identification of an epitope on the dengue virus membrane (M) protein defined by cross-protective monoclonal antibodies: design of an improved epitope sequence based on common determinants present in both envelope (E and M) proteins. Arch Virol 144: 2313–2330.
    1. Kurosu T, Khamlert C, Phanthanawiboon S, Ikuta K, Anantapreecha S (2010) Highly efficient rescue of dengue virus using a co-culture system with mosquito/mammalian cells. Biochem Biophys Res Commun 394: 398–404.
    1. Yenchitsomanus P, Sricharoen P, Jaruthasana I, Pattanakitsakul S, Nitayaphan S, et al. (1996) Rapid detection and identification of dengue viruses by polymerase chain reaction (PCR). Southeast Asian J Trop Med Public Health 27: 228–236.
    1. Shu PY, Chen LK, Chang SF, Yueh YY, Chow L, et al. (2003) Comparison of capture immunoglobulin M (IgM) and IgG Enzyme-linked immunosorbent assay (ELISA) and nonstructural protein NS1 serotype-specific IgG ELISA for differentiation of primary and secondary dengue virus infections. Clin Diagn Lab Immunol 10: 622–630.
    1. Schmittgen TD, Livak KJ (2008) Analyzing real-time PCR data by the comparative CT method. Nat Protoc 3: 1101–1108.
    1. Boonnak K, Slike BM, Donofrio GC, Marovich MA (2013) Human FcγRII cytoplasmic differentially influence antibody-mediated dengue virus infection. J Immunol 190: 5659–5665.
    1. Chareonsirisuthigul T, Kalayanarooj S, Ubol S (2007) Dengue virus (DENV) antibody-dependent enhancement of infection upregulates the production of anti-inflammatory cytokines, but suppresses anti-DENV free radical and pro-inflammatory cytokine production, in THP-1 cells. J Gen Virol 88: 365–375.
    1. Paradkar PN, Ooi EE, Hanson BJ, Gubler DJ, Vasudevan SG (2011) Unfolded protein response (UPR) gene expression during antibody-dependent enhanced infection of cultured monocytes correlates with dengue disease severity. Biosci Rep 31: 221–230.
    1. Modhiran N, Kalayanarooj S, Ubol S (2010) Subversion of innate defenses by the interplay between DENV and pre-existing enhancing antibodies: TLRs signaling collapse. PloS Negl Trop Dis 4: e924.
    1. Moi ML, Lim Ck, Tajima S, Kotaki A, Saijo M, et al. (2011) Dengue virus isolation relying on antibody-dependent enhancement mechanism using FcγR-expressing BHK cells and a monoclonal antibody with infection-enhancing capacity. J Clin Virol 52: 225–230.
    1. Moi ML, Takasaki T, Saijo M, Kurane I (2013) Dengue virus infection-enhancing activity of undiluted sera obtained from patients with secondary dengue virus infection. Trans R Soc Trop Med Hyg 107: 51–58.
    1. Littaua R, Kurane I, Ennis FA (1990) Human IgG Fc receptor II mediates antibody-dependent enhancement of dengue virus infection. J. Immunol. 144: 3183–3186.
    1. Rodrigo WW, Jin X, Blackley SD, Rose RC, Schlesinger J (2006) Differential enhancement of dengue virus immune complex infectivity mediated by signaling-competent and signaling-incompetent human FcγRIA (CD64) or FcγRIIA (CD32). J Virol 80: 10128–10138.
    1. Rodrigo WW, Block OK, Lane C, Sukupolvi-Petty S, Goncalvez AP, et al. (2009) Dengue virus neutralization is modulated by IgG antibody subclass and Fcgamma receptor subtype. Virology 394: 175–182.
    1. Ruangjirachuporn W, Boonpucknavig S, Nimmanitya S (1979) Circulating immune complexes in serum from patients with dengue haemorrhagic fever. Clin Exp Immunol 36: 46–53.
    1. Wang WK, Chen HL, Yang CF, Hsieh SC, Juan CC, et al. (2006) Slower rates of viral load and virus-containing immune complexes in patients with dengue hemorrhagic fever. Clin Infect Dis 43: 1023–30.
    1. Moi ML, Lim CK, Kotaki A, Takasaki T, Kurane I (2011) Detection of higher levels of dengue viremia using FcγR-expressing BHK-21 cells than FcγR-negative cells in secondary infection but not in primary infection. J Infect Dis 203: 1405–1414.
    1. Kou Z, Lim JY, Beltramello M, Quinn M, Chen H, et al. (2011) Human antibodies against dengue enhance dengue viral infectivity without suppressing type I interferon secretion in primary human monocytes. Virology 410: 240–247.
    1. Setthapramote C, Sasaki T, Puiprom O, Limkittikul K, Pitaksajjakul P, et al. (2012) Human monoclonal antibodies to neutralize all dengue virus serotypes using lymphocytes from patients at acute phase of the secondary infection. Biochem Biophy Res Commun 423: 867–872.
    1. Laoprasopwattana K, Libraty DH, Endy TP, Nisalak A, Chunsuttiwat S, et al. (2005) Dengue virus enhancing antibody activity in preillness plasma does not predict subsequent disease severity of viremia in secondary DV infection. J Infect Dis 192: 510–519.
    1. Rodenhuis-Zybert IA, Wilschut J, Smit JM (2010) Dengue virus life cycle: viral and host factors modulating infectivity. Cell Mol Life Sci 67: 2773–2786.

Source: PubMed

3
Abonnere